US20160297889A1 - Pharmaceutical composition for treatment and/or prevention of cancer - Google Patents

Pharmaceutical composition for treatment and/or prevention of cancer Download PDF

Info

Publication number
US20160297889A1
US20160297889A1 US14/910,878 US201414910878A US2016297889A1 US 20160297889 A1 US20160297889 A1 US 20160297889A1 US 201414910878 A US201414910878 A US 201414910878A US 2016297889 A1 US2016297889 A1 US 2016297889A1
Authority
US
United States
Prior art keywords
antibody
seq
chain variable
variable region
cancer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/910,878
Other versions
US9862774B2 (en
Inventor
Fumiyoshi Okano
Takanori Saito
Yoshitaka Minamida
Takayoshi Ido
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Toray Industries Inc
Original Assignee
Toray Industries Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Toray Industries Inc filed Critical Toray Industries Inc
Assigned to TORAY INDUSTRIES, INC. reassignment TORAY INDUSTRIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: OKANO, FUMIYOSHI, IDO, TAKAYOSHI, MINAMIDA, YOSHITAKA, SAITO, TAKANORI
Publication of US20160297889A1 publication Critical patent/US20160297889A1/en
Application granted granted Critical
Publication of US9862774B2 publication Critical patent/US9862774B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • A61K47/48569
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3015Breast
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3023Lung
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/303Liver or Pancreas
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3038Kidney, bladder
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3046Stomach, Intestines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3053Skin, nerves, brain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3061Blood cells
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • C07K16/3069Reproductive system, e.g. ovaria, uterus, testes, prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • C07K2317/732Antibody-dependent cellular cytotoxicity [ADCC]

Definitions

  • the present invention relates to an antibody against CAPRIN-1 or a fragment thereof, and novel pharmaceutical use thereof as a therapeutic and/or preventive agent for a cancer, etc.
  • Cancers are diseases that account for the leading cause of death.
  • the current treatment thereof consists principally of surgical therapy, which may be combined with radiation therapy and/or chemotherapy.
  • novel surgical techniques or the discovery of novel anticancer agents in recent years, the existing treatment of cancers has an insufficiently improved outcome, except for some cancers.
  • antibodies that specifically react with cancers, cancer antigens that are recognized by cytotoxic T cells, genes encoding such cancer antigens, and the like have been identified in recent years, raising expectations on specific cancer therapy targeting the cancer antigens.
  • Cytoplasmic-activation and proliferation-associated protein 1 (CAPRIN-1) has been known as an intracellular protein that is expressed upon activation or cell division of resting normal cells and forms cytoplasmic stress granules with intracellular RNAs to participate in the regulation of transport and translation of mRNAs. This protein has been found to be specifically expressed on the surface of cancer cells and is under study as a target of antibody drugs for cancer treatment (Patent Literatures 1 to 19).
  • An object of the present invention is to produce an antibody that targets CAPRIN-1 specifically expressed on the surface of cancer cells and is superior in antitumor activity to conventional antibodies, and to provide use thereof as a therapeutic and/or preventive agent for a cancer.
  • the present invention provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising an antibody which comprises a heavy chain variable region comprising SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising SEQ ID NOs: 4, 5, and 6 and has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof as an active ingredient.
  • the cancer is breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
  • the antibody is a human antibody, a humanized antibody, a chimeric antibody, a single-chain antibody, or a multispecific antibody (e.g., a bispecific antibody).
  • the antibody against CAPRIN-1 according to the present invention damages cancer cells.
  • the antibody against CAPRIN-1 according to the present invention is useful in the treatment and/or prevention of a cancer.
  • the antibody against a polypeptide of CAPRIN-1 used in the present invention can be evaluated for its antitumor activity, as mentioned later, by examining ex vivo whether or not to exhibit immunocyte-mediated cytotoxic activity against tumor cells expressing the polypeptide or by examining in vivo the inhibition of tumor growth in a cancer-bearing animal.
  • the antibody against CAPRIN-1 according to the present invention may be a monoclonal antibody or a polyclonal antibody and is preferably a monoclonal antibody.
  • the antibody against CAPRIN-1 according to the present invention may be any type of antibody that can exert antitumor activity and includes, for example, recombinant antibodies (e.g., synthetic antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized antibodies, chimeric antibodies, and single-chain antibodies (scFv)), human antibodies, and their antibody fragments (e.g., Fab, F(ab′) 2 , and Fv). These antibodies and fragments thereof can be prepared by methods generally known to those skilled in the art. When a subject is a human, a human antibody or a humanized antibody is desirable for circumventing or suppressing rejection.
  • the phrase “specifically binding to a CAPRIN-1 protein” means that the antibody specifically binds to the CAPRIN-1 protein without substantially binding to other proteins.
  • the subject according to the present invention whose cancer is to be treated and/or prevented is a mammal such as a human, a pet animal, livestock, or a sport animal, and a preferred subject is a human.
  • a proteins or a fragment thereof used as a sensitizing antigen for obtaining the antibody against CAPRIN-1 according to the present invention is not limited by animal species serving as the origin thereof, including humans, dogs, cats, cattle, horses, mice, rats, and chickens. However, it is preferred to select the sensitizing antigen in view of compatibility with parent cells for use in cell fusion. In general, a mammal-derived protein is preferred. Particularly, a human-derived protein is preferred. For example, when CAPRIN-1 is human CAPRIN-1, the human CAPRIN-1 protein, a partial peptide thereof, cells expressing human CAPRIN-1, or the like can be used.
  • nucleotide sequences and amino acid sequences of human CAPRIN-1 and homologs thereof can be obtained, for example, by making an access to GenBank (NCBI, USA) and using an algorithm such as BLAST or FASTA (Karlin and Altschul, Proc. Natl. Acad. Sci. USA, 90: 5873-5877, 1993, and Altschul et al., Nucleic Acids Res. 25: 3389-3402, 1997).
  • the target CAPRIN-1 is a nucleic acid or a protein consisting of a sequence having 70% to 100%, preferably 80% to 100%, more preferably 90% to 100%, further preferably 95% to 100%, for example, 97% to 100%, 98% to 100%, 99% to 100%, or 99.5% to 100% sequence identity to the nucleotide sequence or amino acid sequence of the ORF or mature portion of the reference (the amino acid sequences of SEQ ID NO: 17 and SEQ ID NO: 19 compared with each other differ in amino acid residues at and following position 690).
  • % sequence identity means a percentage (%) of the number of identical amino acids (or bases) to the total number (including the number of gaps) of amino acids (or bases) when two sequences are aligned such that the maximum degree of similarity or identity can be achieved with or without introduced gaps.
  • the fragment of the CAPRIN-1 protein has a length ranging from the amino acid length of an epitope (antigenic determinant), which is the smallest unit recognized by the antibody, to less than the full length of the protein.
  • the epitope refers to a polypeptide fragment having antigenicity or immunogenicity in mammals, preferably humans. Its smallest unit consists of approximately 7 to 12 amino acids, for example, 8 to 11 amino acids.
  • a polypeptide fragment comprising the aforementioned human CAPRIN-1 protein or a partial peptide thereof can be synthesized according to a chemical synthesis method, for example, an Fmoc (fluorenylmethyloxycarbonyl) or tBoc (t-butyloxycarbonyl) method (Seikagaku Jikken Koza 1 (Biochemical Experimentation Course 1 in English), the Japanese Biochemical Society ed., Protein Chemistry IV. Chemical Modification and Peptide Synthesis, Tokyo Kagaku Dojin Co., Ltd. (Japan), 1981).
  • the human CAPRIN-1 protein or the polypeptide fragment can be synthesized by a routine method using various commercially available peptide synthesizers.
  • a polynucleotide encoding the polypeptide is prepared using a genetic engineering approach known in the art (Sambrook et al., Molecular Cloning, the 2nd edition. Current Protocols in Molecular Biology (1989), Cold Spring Harbor Laboratory Press: Ausubel et al., Short Protocols in Molecular Biology, the 3rd edition, A compendium of Methods from Current Protocols in Molecular Biology (1995). John Wiley & Sons: etc.), and this polynucleotide is incorporated into expression vectors, which are then transferred to host cells so that the polypeptide is produced in the host cells. In this way, the human CAPRIN-1 protein of interest or the polypeptide fragment thereof can be obtained.
  • the polynucleotide encoding the polypeptide can be readily prepared by a genetic engineering approach known in the art or a routine method using a commercially available nucleic acid synthesizer.
  • a DNA comprising the nucleotide sequence of human CAPRIN-1 gene can be prepared by PCR using a human chromosomal DNA or cDNA library as a template and a pair of primers designed so as to be able to amplify the nucleotide sequence.
  • Reaction conditions for this PCR can be appropriately determined. Examples of the conditions can include, but are not limited to, 30 cycles each involving reaction steps consisting of 94° C. for 30 seconds (denaturation), 55° C. for 30 seconds to 1 minute (annealing), and 72° C.
  • thermostable DNA polymerase e.g., Taq polymerase or Pfu polymerase
  • Mg 2+ -containing PCR buffer e.g., a Mg 2+ -containing PCR buffer
  • the PCR approach, conditions, etc. are described in, for example, Ausubel et al., Short Protocols in Molecular Biology, the 3rd edition, A Compendium of Methods from Current Protocols in Molecular Biology (1995), John Wiley & Sons (particularly, Chapter 15).
  • probes or primers can be prepared on the basis of information on the nucleotide sequence of the CAPRIN-1 gene and the amino acid sequence of the CAPRIN-1 protein, and used in the screening of, for example, a human cDNA library, to isolate the desired DNA. It is preferred that the cDNA library should be produced from cells, organs, or tissues expressing the CAPRIN-1 protein.
  • cells or tissues include cells or tissues derived from the testis as well as from cancers or tumors such as leukemia, breast cancer, lymphoma, brain tumor, lung cancer, pancreatic cancer, colorectal cancer, kidney cancer, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, sarcoma, mastocytoma, liver cancer, gallbladder cancer, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, and head and neck cancer.
  • cancers or tumors such as leukemia, breast cancer, lymphoma, brain tumor, lung cancer, pancreatic cancer, colorectal cancer, kidney cancer, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, sarcoma, mastocytoma, liver cancer, gallbladder cancer, mela
  • the host cells to receive the expression vectors may be any cell capable of expressing the polypeptide.
  • prokaryotic cells include, but are not limited to, E. coli .
  • eukaryotic cells include, but are not limited to: mammalian cells such as monkey kidney cells COS 1 and Chinese hamster ovary cells CHO; a human embryonic kidney cell line HEK293; a mouse embryonic skin cell line NIH3T3; yeast cells such as budding yeast and fission yeast cells; silkworm cells; and Xenopus egg cells.
  • expression vectors having an origin that permits replication in the prokaryotic cells are used as the expression vectors.
  • Examples of expression vectors for E. coli can include pUC series, pBluescript II, pET expression systems, and pGEX expression systems.
  • the DNA encoding the polypeptide is incorporated into such expression vectors, and prokaryotic host cells can be transformed with these vectors, followed by the culture of the obtained transformants so that the polypeptide encoded by the DNA is expressed in the prokaryotic host cells.
  • the polypeptide may be expressed as a fusion protein with another protein.
  • expression vectors for eukaryotic cells having a promoter, a splicing region, a poly(A) addition site, etc. are used as the expression vectors.
  • expression vectors can include pKA1, pCDM8, pSVK3, pMSG, pSVL, pBK-CMV, pBK-RSV, EBV, pRS, pcDNA3, and pYES2 vectors.
  • the DNA encoding the polypeptide is incorporated into such expression vectors, and eukaryotic host cells can be transformed with these vectors, followed by the culture of the obtained transformants so that the polypeptide encoded by the DNA is expressed in the eukaryotic host cells.
  • expression vectors such as pIND/V5-His, pFLAG-CMV-2, pEGFP-N1, or pEGFP-C1
  • the polypeptide can be expressed as various fusion proteins tagged with His tag (e.g., (His) 6 to (His) 10 ), FLAG tag, myc tag. HA tag, GFP, or the like.
  • the transfer of the expression vectors to the host cells can employ a well-known method such as electroporation, a calcium phosphate method, a liposome method, a DEAE dextran method, microinjection, viral infection, lipofection, or binding with cell-penetrating peptides.
  • Separation treatments known in the art can be carried out in combination for isolating and purifying the polypeptide of interest from the host cells.
  • a denaturant e.g., urea
  • a surfactant e.g., ultrasonication, enzymatic digestion, salting-out, solvent fractionation and precipitation
  • dialysis centrifugation, ultrafiltration, gel filtration, SDS-PAGE, isoelectric focusing electrophoresis, ion-exchange chromatography, hydrophobic chromatography, affinity chromatography, and reverse-phase chromatography.
  • the antigen thus prepared can be used as a sensitizing antigen as mentioned later for producing the antibody according to the present invention.
  • Antibodies are usually heteromultimeric glycoproteins each comprising at least two heavy chains and two light chains.
  • the immunoglobulins, except for IgM, are heterotetrameric glycoproteins of approximately 150 kDa each composed of two identical light (L) chains and two identical heavy (H) chains.
  • L light
  • H heavy
  • each light chain is connected to a heavy chain via a single covalent disulfide bond, though the number of disulfide bonds varies among heavy chains of different immunoglobulin isotypes.
  • Each heavy chain and light chain also have an intrachain disulfide bond.
  • Each heavy chain has a variable domain (VH region) at one end, followed by a series of constant regions.
  • Each light chain has a variable domain (VL region) at one end and has a single constant region at the other end.
  • the light chain constant region is aligned with the first heavy chain constant region, while the light chain variable domain is aligned with the heavy chain variable domain.
  • Particular regions called complementarity-determining regions (CDRs) in the antibody variable domains exhibit specific variability and impart binding specificity to the antibody. Moietys relatively conserved in the variable regions are called framework regions (FRs).
  • the complete heavy chain and light chain variable domains each has a structure in which four framework regions are connected via three complementarity-determining regions, i.e., a structure in which FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 are connected in this order from the N-terminus.
  • CDRH1, CDRH2, and CDRH3 are called CDRH1, CDRH2, and CDRH3 in this order from the N-terminus thereof.
  • CDRH3 is most important for the binding specificity of the antibody for its antigen.
  • CDRs in each chain are kept close to each other by the framework regions and contribute to the formation of an antigen-binding site in the antibody, together with the complementarity-determining regions derived from the other chain.
  • the constant regions do not directly contribute to antibody-antigen binding, but exhibit various effector functions, for example, involvement in antibody-dependent cellular cytotoxicity (ADCC), phagocytosis (ADCP) mediated by binding to an Fc ⁇ receptor, half-life/clearance rates mediated by a neonatal Fc receptor (FcRn), and complement-dependent cytotoxicity (CDC) mediated by a Clq component in the complement cascade.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP phagocytosis
  • FcRn neonatal Fc receptor
  • CDC complement-dependent cytotoxicity
  • the anti-CAPRIN-1 antibody according to the present invention means an antibody having immunological reactivity with a full-length CAPRIN-1 protein or a fragment thereof.
  • the “immunological reactivity” means the property of the antibody binding to the CAPRIN-1 antigen (a full-length CAPRIN-1 protein or a partial polypeptide thereof) in vivo. Via such binding to CAPRIN-1, the antibody of the present invention exerts the function of damaging (e.g., killing, suppressing, or regressing) tumor cells.
  • the antibody of the present invention can damage tumor, for example, breast cancer, kidney cancer, pancreatic cancer, colorectal cancer (e.g., colon cancer), lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer.
  • Ewing's tumor Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer through binding to the CAPRIN-1 protein.
  • the antibody of the present invention is not particularly limited, preferably, as long as the antibody is a monoclonal antibody.
  • the antibody of the present invention includes synthetic antibodies, multispecific antibodies (e.g., diabodies and triabodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain antibodies, antibody fragments (e.g., Fab, F(ab′) 2 , and Fv), and the like.
  • the antibody is an immunoglobulin molecule of any class, for example, IgG, IgE, IgM, IgA, IgD, or IgY, or of any subclass, for example, IgG1, IgG2, IgG3, IgG4, IgA1, or IgA2.
  • the antibody may be further modified by deglycosylation, acetylation, formylation, amidation, phosphorylation, PEGylation, or the like, in addition to glycosylation.
  • a breast cancer cell line SK-BR-3 expressing CAPRIN-1 is administered to a mouse for immunization.
  • the spleen is extracted from this mouse. After separation of spleen cells, the cells are fused with mouse myeloma cells.
  • Clones producing antibodies having a cancer cell growth inhibitory effect are selected from among the obtained fusion cells (hybridomas).
  • the hybridomas producing monoclonal antibodies having a cancer cell growth inhibitory effect are isolated, and these hybridomas are cultured.
  • the antibody of the present invention can be prepared by purification from the culture supernatant according to a general affinity purification method.
  • the monoclonal antibody-producing hybridomas may be prepared, for example, as follows: first, animals are immunized with the sensitizing antigen according to a method known in the art. This immunization method generally involves intraperitoneally or subcutaneously injecting the sensitizing antigen to mammals. Specifically, the sensitizing antigen diluted with or suspended in PBS (phosphate-buffered saline), physiological saline, or the like into an appropriate amount is mixed, if desired, with an appropriate amount of a conventional adjuvant, for example, a Freund's complete adjuvant. After emulsification, the resulting emulsion is administered to each mammal several times every 4 to 21 days. Alternatively, an appropriate carrier may be used for the immunization with the sensitizing antigen.
  • PBS phosphate-buffered saline
  • physiological saline physiological saline
  • an appropriate carrier may be used for the immunization with the sensitizing antigen.
  • immunocytes are collected from the mammal and subjected to cell fusion.
  • Preferred examples of the immunocytes particularly include spleen cells.
  • Mammalian myeloma cells are used as partner parent cells to be fused with the immunocytes.
  • Various cell lines known in the art for example, P3U1 (P3-X63Ag8U1), P3 (P3x63Ag8.653) (J. Immunol. (1979) 123, 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (Kohler. G. and Milstein. C. Eur. J. Immunol. (1976) 6, 511-519), MPC-1 (Margulies. D. H. et al., Cell (1976) 8, 405-415), SP2/0 (Shulman. M.
  • the cell fusion between the immunocytes and the myeloma cells can be carried out basically according to a method known in the art, for example, the method of Kohler and Milstein (Kohler, G. and Milstein, C. Methods Enzymol. (1981) 73, 3-46).
  • the cell fusion is carried out, for example, in the presence of a cell fusion promoter in a conventional nutrient medium.
  • a cell fusion promoter for example, polyethylene glycol (PEG) or hemagglutinating virus of Japan (HVJ) is used as the fusion promoter.
  • PEG polyethylene glycol
  • HVJ hemagglutinating virus of Japan
  • an auxiliary such as dimethyl sulfoxide may be further added for use in order to enhance fusion efficiency.
  • the ratio between the immunocytes and the myeloma cells used can be arbitrarily set.
  • an RPMII640 medium or a MEM medium suitable for the growth of the myeloma cell lines, or a conventional medium for use in this type of cell culture can be used as the medium for use in the cell fusion.
  • a serum supplement such as fetal calf serum (FCS) may be used in combination with the medium.
  • FCS fetal calf serum
  • the immunocytes and the myeloma cells are well mixed in a predetermined amount of the medium.
  • a PEG solution average molecular weight: for example, approximately 1000 to 6000
  • preheated to approximately 37° C. is usually added to the mixture at a concentration of 30 to 60% (w/v) and mixed therewith to form the hybridomas of interest.
  • it is preferred to remove cell fusion agents or the like unfavorable for the growth of the hybridomas by repeating the procedures of sequentially adding an appropriate medium and removing the supernatant by centrifugation.
  • the hybridomas thus obtained are cultured in a conventional selective medium, for example, a HAT medium (medium containing hypoxanthine, aminopterin, and thymidine) for selection.
  • a HAT medium medium containing hypoxanthine, aminopterin, and thymidine
  • This culture in the HAT medium is continued for a period (usually, several days to several weeks) sufficient for the death of cells (non-fused cells) other than the hybridomas of interest.
  • hybridomas producing the antibody of interest are screened for and cloned as single clones by a conventional limiting dilution method.
  • hybridomas producing human antibodies having the desired activity may be obtained by sensitizing human lymphocytes, for example, EB virus-infected human lymphocytes, with the protein, protein-expressing cells, or lysates thereof in vitro and fusing the sensitized lymphocytes with human-derived myeloma cells capable of dividing permanently, for example, U266 (Registration No. TIB196).
  • the monoclonal antibody-producing hybridomas thus prepared can be subcultured in a conventional medium and can also be stored for a long period in liquid nitrogen.
  • the desired antigen or cells expressing the desired antigen are used as a sensitizing antigen in immunization according to a conventional immunization method.
  • the obtained immunocytes are fused with parent cells known in the art according to a conventional cell fusion method.
  • Monoclonal antibody-producing cells can be screened for by a conventional screening method to prepare the antibody of interest.
  • the antigen can be prepared according to, for example, a method using animal cells (JP Patent Publication (Kohyo) No. 2007-530068 A (2007)) or a method using baculovirus (e.g., International Publication No. WO98/46777).
  • this antigen can be bound to an immunogenic macromolecule such as albumin for immunization.
  • the antigen may be administered together with an adjuvant for immunization.
  • the antibody of the present invention may be obtained as a gene recombinant antibody produced using a gene recombination technique which involves: cloning a gene of the antibody from a hybridoma: incorporating the antibody gene into appropriate vectors, and transferring the vectors into hosts (see, e.g., Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990).
  • V region variable region
  • the DNAs are ligated with DNAs encoding the desired antibody constant regions (C regions).
  • the resulting ligation products are incorporated into expression vectors.
  • the antibody V region-encoding DNAs may be incorporated into expression vectors containing antibody C region DNAs. These DNAs are incorporated into the expression vectors so as to be expressed under the control of expression control regions, for example, an enhancer and a promoter.
  • host cells can be transformed with the resulting expression vectors and allowed to express the antibody.
  • the anti-CAPRIN-1 antibody of the present invention is preferably a monoclonal antibody.
  • the monoclonal antibody includes human monoclonal antibodies, non-human animal monoclonal antibodies (e.g., mouse, rat, rabbit, and chicken monoclonal antibodies), chimeric monoclonal antibodies, and the like.
  • the monoclonal antibody may be prepared by culturing hybridomas obtained by the fusion between spleen cells from non-human mammals (e.g., mice, human antibody-producing mice, chickens, or rabbits) immunized with the CAPRIN-1 protein or a fragment thereof and myeloma cells.
  • the chimeric antibody is an antibody prepared from a combination of sequences derived from different animals and is, for example, an antibody composed of heavy chain and light chain variable regions of a mouse antibody and heavy chain and light chain constant regions of a human antibody.
  • the chimeric antibody can be prepared using a method known in the art and is obtained, for example by: ligating DNAs encoding the antibody V regions with DNAs encoding the human antibody C regions; incorporating the resulting ligation products into expression vectors; and transferring the vectors into hosts for antibody production.
  • a plurality of humanized monoclonal antibodies and a human-rabbit chimeric monoclonal antibody were prepared and confirmed to have a strong antitumor effect.
  • All of these monoclonal antibodies have a heavy chain variable region (VH region) comprising CDR1 represented by the amino acid sequence of SEQ ID NO: 1, CDR2 represented by the amino acid sequence of SEQ ID NO: 2, and CDR3 represented by the amino acid sequence of SEQ ID NO: 3, and a light chain variable region (VL region) comprising CDR1 represented by the amino acid sequence of SEQ ID NO: 4, CDR2 represented by the amino acid sequence of SEQ ID NO: 5, and CDR3 represented by the amino acid sequence of SEQ ID NO: 6.
  • VH region heavy chain variable region
  • CDR1 represented by the amino acid sequence of SEQ ID NO: 1
  • CDR2 represented by the amino acid sequence of SEQ ID NO: 2
  • CDR3 represented by the amino acid sequence of SEQ ID NO: 3
  • VL region light chain variable region
  • These monoclonal antibodies include humanized antibody #0 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 11, humanized antibody #1 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 11, humanized antibody #2 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 12, humanized antibody #3 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 13, humanized antibody #4 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 12, humanized antibody #5 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence
  • the humanized antibody also called reshaped human antibody, is an engineered antibody.
  • the humanized antibody is constructed by grafting complementarity-determining regions of a human antibody with complementarity-determining regions of an antibody derived from an immunized animal. A general gene recombination approach therefor is also known.
  • DNA sequences designed so as to link complementarity-determining regions of, for example, a mouse, rabbit, or chicken antibody, and framework regions of a human antibody are synthesized by PCR from several prepared oligonucleotides having terminal portions overlapping with each other. The obtained DNAs are ligated with DNAs encoding human antibody constant regions. Subsequently, the resulting ligation products are incorporated into expression vectors, which are then transferred to hosts for antibody production to obtain the antibody of interest (see European Patent Application Publication No. EP239400 and International Publication No. WO96/02576).
  • the framework regions of a human antibody connected via the complementarity-determining regions are selected such that the complementarity-determining regions form a favorable antigen-binding site.
  • amino acids in the framework regions of antibody variable regions may be substituted such that the complementarity-determining regions of the resulting reshaped human antibody form an appropriate antigen-binding site (Sato K. et al., Cancer Research 1993, 53: 851-856).
  • these framework regions may be replaced with framework regions derived from various human antibodies (see International Publication No. WO99/51743).
  • variable regions e.g., FRs
  • constant regions e.g., amino acids in variable regions or constant regions may be substituted, for example, by other amino acids.
  • the amino acid substitution is the substitution of one or more, for example, less than 15, less than 10, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less amino acids, preferably 1 to 9 amino acids.
  • the substituted antibody should be functionally equivalent to an unsubstituted antibody.
  • the phrase “functionally equivalent” means that an antibody concerned has biological or biochemical activity similar to that of the antibody of the present invention, specifically, the antibody concerned has the function of damaging tumor and essentially causes no rejection when applied to humans, for example.
  • Examples of such activity can include cell growth inhibitory activity and binding activity.
  • An amino acid substitution method which involves introducing a mutation into a polypeptide is well known to those skilled in the art as a method for preparing a polypeptide functionally equivalent to a certain polypeptide.
  • those skilled in the art can appropriately introduce amino acid substitution into the antibody of the present invention using site-directed mutagenesis (Hashimoto-Gotoh, T. et al., (1995) Gene 152, 271-275; Zoller, M J., and Smith, M. (1983) Methods Enzymol. 100, 468-500; Kramer, W. et al., (1984) Nucleic Acids Res. 12, 9441-9456: Kramer, W. and Fritz, H J., (1987) Methods Enzymol.
  • the substitution is desirably conservative amino acid substitution.
  • the conservative amino acid substitution is the substitution between amino acids similar in properties such as charge, side chains, polarity, and aromaticity.
  • the amino acids can be classified in terms of similar properties into, for example: basic amino acids (arginine, lysine, and histidine); acidic amino acids (aspartic acid and glutamic acid); uncharged polar amino acids (glycine, asparagine, glutamine, serine, threonine, cysteine, and tyrosine); nonpolar amino acids (leucine, isoleucine, alanine, valine, proline, phenylalanine, tryptophan, and methionine): branched amino acids (leucine, valine, and isoleucine); and aromatic amino acids (phenylalanine, tyrosine, tryptophan, and histidine).
  • modified antibodies can include antibodies bound with various molecules such as polyethylene glycol (PEG).
  • PEG polyethylene glycol
  • the substance to be bound is not limited.
  • the obtained antibody can be chemically modified. A method therefor has already been established in the art.
  • the antibody that recognizes a CAPRIN-1 protein or a CAPRIN-1 fragment polypeptide can be obtained by a method generally known to those skilled in the art.
  • the antibody can be obtained by, for example, a method which involves determining an epitope of the CAPRIN-1 protein recognized by an anti-CAPRIN-1 antibody by a conventional method (e.g., epitope mapping or an epitope identification method mentioned later) and preparing an antibody using a polypeptide having an amino acid sequence contained in the epitope as an immunogen, or a method which involves determining an epitope for an antibody prepared by a conventional method and selecting an antibody that recognizes the same epitope as that for an anti-CAPRIN-1 antibody.
  • the antibody of the present invention is an antibody having immunological reactivity with CAPRIN-1, an antibody that specifically recognizes CAPRIN-1, or an antibody that specifically binds to CAPRIN-1 and exhibits cytotoxic activity against a cancer or a tumor growth inhibitory effect. It is preferred that the antibody should be an antibody having a structure so as to cause little or no rejection in recipient animals. Examples of such antibodies include human antibodies, humanized antibodies, chimeric antibodies (e.g., human-rabbit chimeric antibodies), single-chain antibodies, and bispecific antibodies when the recipient animals are humans.
  • These antibodies are recombinant antibodies having heavy chain and light chain variable regions derived from a human antibody, having heavy chain and light chain variable regions comprising complementarity-determining regions (CDR1, CDR2, and CDR3) derived from a non-human animal antibody and framework regions (FR1, FR2, FR3, and FR4) derived from a human antibody, or having heavy chain and light chain variable regions derived from a non-human animal antibody and heavy chain and light chain constant regions derived from a human antibody.
  • CDR1, CDR2, and CDR3 complementarity-determining regions
  • FR1, FR2, FR3, and FR4 non-human animal antibody and framework regions
  • Preferred antibodies are the former two antibodies.
  • recombinant antibodies can be prepared as follows: a DNA encoding the monoclonal antibody (e.g., a human, mouse, rat, rabbit, or chicken monoclonal antibody) against human CAPRIN-1 is cloned from antibody-producing cells such as hybridomas, and this is used as a template in RT-PCR or the like to prepare DNAs encoding the light chain and heavy chain variable regions of the antibody.
  • a DNA encoding the monoclonal antibody e.g., a human, mouse, rat, rabbit, or chicken monoclonal antibody
  • human CAPRIN-1 e.g., human, mouse, rat, rabbit, or chicken monoclonal antibody
  • the respective sequences of the light chain and heavy chain variable regions, the respective sequences of CDR1, CDR2, and CDR3 in each region, or the respective sequences of FR1, FR2, FR3, and FR4 in each region can be determined on the basis of, for example, the Kabat EU numbering system (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institute of Health, Bethesda, Md. (1991)).
  • Such a DNA encoding each of these variable regions or a DNA encoding each complementarity-determining region is further prepared using a gene recombination technique (Sambrook et al., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)) or a DNA synthesizer.
  • the human monoclonal antibody-producing hybridomas can be prepared by immunizing human antibody-producing animals (e.g., mice) with human CAPRIN-1 and then fusing spleen cells excised from the immunized animals with myeloma cells.
  • DNAs encoding human antibody-derived light chain or heavy chain variable and constant regions are prepared, if necessary, using a gene recombination technique or a DNA synthesizer.
  • the CDR coding sequences in the DNA encoding the human antibody-derived light chain or heavy chain variable region can be substituted by corresponding CDR coding sequences of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody to thereby prepare a humanized antibody-encoding DNA.
  • a humanized antibody in which CDR coding sequences derived from a human antibody is substituted by corresponding CDR coding sequences derived from a mouse antibody each variable region is constituted by human FR1, mouse CDR1, human FR2, mouse CDR2, human FR3, mouse CDR4, and human FR4 in this order from the N-terminus.
  • the DNA encoding the light chain or heavy chain variable region of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody can be ligated with the DNA encoding a light chain or heavy chain constant region derived from a human antibody to prepare a chimeric antibody-encoding DNA.
  • a non-human animal e.g., mouse, rat, rabbit, or chicken
  • this antibody refers to an antibody comprising a heavy chain variable region and a light chain variable region linearly linked via a linker.
  • a DNA encoding the single-chain antibody can be prepared by ligating a DNA encoding the heavy chain variable region, a DNA encoding the linker, and a DNA encoding the light chain variable region.
  • both of the heavy chain variable region and the light chain variable region are derived from a human antibody or derived from a human antibody having complementarity-determining regions alone substituted by complementarity-determining regions of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody.
  • the linker consists of 12 to 19 amino acids. Examples thereof include (G 4 S) 3 consisting of 15 amino acids (G.-B. Kim et al., Protein Engineering Design and Selection 2007, 20 (9): 425-432).
  • this antibody refers to an antibody capable of specifically binding to two different epitopes.
  • a DNA encoding the bispecific antibody can be prepared by ligating, for example, a DNA encoding heavy chain variable region A, a DNA encoding light chain variable region B, a DNA encoding heavy chain variable region B, and a DNA encoding light chain variable region A in this order (provided that the DNA encoding a light chain variable region B and the DNA encoding a heavy chain variable region B are ligated via a DNA encoding a linker as described above).
  • all of the heavy chain variable regions and the light chain variable regions are derived from a human antibody or derived from a human antibody having complementarity-determining regions alone substituted by complementarity-determining regions of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody.
  • a non-human animal e.g., mouse, rat, rabbit, or chicken
  • the recombinant DNAs thus prepared can be incorporated into one or more appropriate vectors, which are then transferred to host cells (e.g., mammalian cells, yeast cells, and insect cells) so that the DNAs are (co)expressed to produce the recombinant antibody of interest (P. J. Delves., ANTIBODY PRODUCTION ESSENTIAL TECHNIQUES., 1997 WILEY: P. Shepherd and C. Dean., Monoclonal Antibodies., 2000 OXFORD UNIVERSITY PRESS; J. W. Goding., Monoclonal Antibodies: principles and practice., 1993 ACADEMIC PRESS).
  • host cells e.g., mammalian cells, yeast cells, and insect cells
  • Examples of the antibody of the present invention prepared by any of the methods described above include the following antibodies (a) to (1) comprising a heavy chain variable region comprising SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising SEQ ID NOs: 4, 5, and 6, obtained in Examples mentioned later:
  • amino acid sequences represented by SEQ ID NOs: 1, 2, and 3 correspond to CDR1, CDR2, and CDR3, respectively, of the heavy chain variable region of the rabbit antibody
  • amino acid sequences represented by SEQ ID NOs: 4, 5, and 6 correspond to CDR1, CDR2, and CDR3, respectively, of the light chain variable region of the rabbit antibody.
  • the humanized antibody, the chimeric antibody, the single-chain antibody, or the bispecific antibody of the present invention is, for example, any of the following antibodies (i) to (xiv):
  • an antibody comprising a heavy chain variable region comprising the amino acid sequences of SEQ ID NOs: 1, 2, and 3 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof, and a light chain variable region comprising the amino acid sequences of SEQ ID NOs: 4, 5, and 6 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequences of SEQ ID NOs: 1, 2, and 3 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequences of SEQ ID NOs: 4, 5, and 6 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence.
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 13 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 13 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 20 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 21 and a light chain constant region comprising a human antibody-derived amino acid sequence.
  • sequences of framework regions of the human antibody heavy chain and light chain constant and variable regions are available from, for example, NCBI (USA; GenBank, UniGene, etc.). For example, the following sequences can be referred to: Registration No. J00228 for a human IgG1 heavy chain constant region; Registration No. J00230 for a human IgG2 heavy chain constant region; Registration No. X03604 for a human IgG3 heavy chain constant region: Registration No. K01316 for a human IgG4 heavy chain constant region; Registration Nos. V00557, X64135, X64133, etc., for a human light chain ⁇ constant region: and Registration Nos. X64132. X64134, etc., for a human light chain ⁇ constant region.
  • these antibodies have cytotoxic activity and can thereby exert an antitumor effect (or antitumor activity).
  • the aforementioned antibodies may each have the substitution, deletion, or addition of one or several amino acids in a complementarity-determining region sequence, a framework region sequence, and/or a constant region sequence, as long as the resulting antibody has such specificity that it can specifically recognize CAPRIN-1.
  • the term “several” means preferably 1 to 9.
  • the affinity constant Ka (k on /k off ) of the antibody of the present invention for a CAPRIN-1 protein or a fragment thereof is preferably at least 5 ⁇ 10 8 M ⁇ 1 , at least 10 9 M ⁇ 1 , at least 5 ⁇ 10 9 M ⁇ 1 , at least 10 10 M ⁇ 1 , at least 5 ⁇ 10 10 M ⁇ 1 , at least 10 11 M ⁇ 1 , at least 5 ⁇ 10 11 M ⁇ 1 , at least 10 12 M ⁇ 1 , at least 10 13 M ⁇ 1 , or at least 10 14 M ⁇ 1 ,
  • ADCC antibody-dependent cellular cytotoxicity
  • Such an antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain attached to the heavy chain constant region according to the present invention may be used alone or may be a composition with a fucosylated antibody. It is preferred that the antibody composition should be composed mainly of the antibody lacking fucose.
  • the antibody in which one or several amino acids in the heavy chain constant region are substituted can be prepared with reference to, for example, International Publication No. WO2004/063351, International Publication No. WO2011/120135, U.S. Pat. No. 8,388,955, International Publication No. WO2011/005481, U.S. Pat. No. 6,737,056, and International Publication No. WO2005/063351.
  • the antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain in the heavy chain constant region, or cells producing the antibody can be prepared with reference to, for example, U.S. Pat. No. 6,602,684, European Patent No. 1914244, and U.S. Pat. No.
  • composition of the antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain attached to the heavy chain constant region, and the fucosylated antibody, or cells producing the composition can be prepared with reference to, for example, U.S. Pat. No. 8,642,292.
  • the antibody of the present invention can be conjugated with an antitumor agent.
  • the conjugation of the antibody with the antitumor agent can be carried out via a spacer having a group (e.g., a succinimidyl group, a formyl group, a 2-pyridyldithio group, a maleimidyl group, an alkoxycarbonyl group, or a hydroxy group) reactive with an amino group, a carboxyl group, a hydroxy group, a thiol group, or the like.
  • a group e.g., a succinimidyl group, a formyl group, a 2-pyridyldithio group, a maleimidyl group, an alkoxycarbonyl group, or a hydroxy group
  • antitumor agent examples include the following antitumor agents publicly known in literatures, etc.: paclitaxel, doxorubicin, daunorubicin, cyclophosphamide, methotrexate, 5-fluorouracil, thiotepa, busulfan, improsulfan, piposulfan, benzodopa, carboquone, meturedopa, uredopa, altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, trimethylolomelamine, bullatacin, bullatacinone, camptothecin, bryostatin, callystatin, cryptophycin 1, cryptophycin 8, dolastatin, duocarmycin, eleutherobin, pancratistatin, sarcodictyin, spongistatin, chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifo
  • a method for evaluating whether to exert antitumor activity can involve, for example, for the mouse-derived anti-CAPRIN-1 antibody, reacting a drug-attached secondary antibody binding to a mouse antibody together therewith to evaluate ex vivo the antitumor effect on human cancer cells.
  • This evaluation can be conducted using, for example, an anti-human IgG antibody (Hum-ZAP (Advanced Targeting Systems, Inc.)), which is a second immunotoxin bound with saporin.
  • the antibody of the present invention can be administered in combination with an antitumor agent to thereby produce a higher therapeutic effect.
  • This approach is adaptable to a patient with a CAPRIN-1-expressing cancer either before or after surgical treatment.
  • This approach can be applied, particularly after surgery, to a CAPRIN-1-expressing cancer, which has been treated conventionally with an antitumor agent alone, to produce higher prevention of cancer recurrence or prolongation of survival time.
  • any of the antitumor agents described above can be used as the antitumor agent for use in the combined administration with the antibody of the present invention.
  • cyclophosphamide, paclitaxel, docetaxel, or vinorelbine is preferably used.
  • the antibody of the present invention may be bound to a radioisotope publicly known in literatures, etc., such as 211 At, 131 I, 125 I, 90 Y, 186 Re, 188 Re, 153 SM, 212 Bi, 32 P, 175 Lu, 176 Lu, 89 Sr, 64 Cu, or 111 In (Hideo Saji, YAKUGAKU ZASSHI 128 (3) 323-332, 8 (2008), Jpn).
  • a radioisotope effective for the treatment or diagnosis of tumor is desirable.
  • Such a radioisotope is also included in the antitumor agent according to the present invention.
  • the antitumor effect of the anti-CAPRIN-1 antibody used in the present invention on CAPRIN-1-expressing cancer cells is considered to take place under the following mechanism or the like: the antibody-dependent cellular cytotoxicity (ADCC) of effector cells against the CAPRIN-1-expressing cells mentioned above and the antibody-dependent cellular phagocytosis (ADCP) of the CAPRIN-1-expressing cells.
  • ADCC antibody-dependent cellular cytotoxicity
  • ADCP antibody-dependent cellular phagocytosis
  • the activity of the anti-CAPRIN-1 antibody used in the present invention can be evaluated, as specifically shown below in Examples, by measuring ex vivo the ADCC activity or the ADCP activity against CAPRIN-1-expressing cancer cells.
  • the anti-CAPRIN-1 antibody used in the present invention binds to the CAPRIN-1 protein on cancer cells and exhibits an antitumor effect through the activity or the like.
  • the anti-CAPRIN-1 antibody of the present invention is presumably useful in the treatment or prevention of a cancer.
  • the present invention provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising the anti-CAPRIN-1 antibody as an active ingredient.
  • the anti-CAPRIN-1 antibody used for the purpose of administration to human bodies (antibody therapy) is preferably a human antibody or a humanized antibody for reducing immunogenicity.
  • an anti-CAPRIN-1 antibody with higher binding affinity for the CAPRIN-1 protein on the surface of cancer cells exerts stronger antitumor activity.
  • the antibody of the present invention has high binding affinity for the CAPRIN-1 protein and can therefore be expected to have a stronger antitumor effect. Accordingly, the antibody of the present invention is adaptable to a pharmaceutical composition intended for the treatment and/or prevention of a cancer.
  • Such high binding affinity of the antibody of the present invention is preferably at least 5 ⁇ 10 8 M ⁇ 1 , at least 10 9 M ⁇ 1 , at least 5 ⁇ 10 9 M ⁇ 1 , at least 10 10 M ⁇ 1 , at least 5 ⁇ 10 10 M ⁇ 1 , at least 10 11 M ⁇ 1 , at least 5 ⁇ 10 11 M ⁇ 1 , at least 10 12 M ⁇ 1 , at least 10 13 M ⁇ 1 , or at least 10 14 M
  • 1 in terms of an association constant (affinity constant) Ka (k on /k off ), as described above.
  • the ability of the antibody to bind to CAPRIN-1 can be determined by use of binding assay using, for example, ELISA, Western blot, immunofluorescence, and flow cytometry analysis, as described in Examples.
  • the antibody that recognizes CAPRIN-1 can be used in immunohistochemistry by a method well known to those skilled in the art.
  • the antibody that recognizes CAPRIN-1 can be tested for its reactivity with CAPRIN-1, for example, using a paraformaldehyde- or acetone-fixed frozen section or a paraformaldehyde-fixed and paraffin-embedded section of a tissue obtained from a patient during surgical treatment or a tissue obtained from an animal carrying a xenograft tissue inoculated with a cell line expressing CAPRIN-1 either spontaneously or after transfection.
  • the antibody reactive with CAPRIN-1 can be stained by various methods.
  • the antibody can be visualized through reaction with a horseradish peroxidase-conjugated goat anti-mouse antibody, goat anti-rabbit antibody, or goat anti-chicken antibody.
  • the target of the pharmaceutical composition for the treatment and/or prevention of a cancer of the present invention is not particularly limited as long as the target is cancer (cells) expressing a CAPRIN-1 gene.
  • tumor and cancer used in the present specification mean malignant neoplasm and are used interchangeably with each other.
  • the cancer targeted in the present invention may be any cancer expressing the CAPRIN-1 protein on the cell membrane surface.
  • the cancer is preferably breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer as mentioned above.
  • examples of these cancers include, but are not limited to, breast adenocarcinoma, complex-type breast adenocarcinoma, malignant mixed tumor of the mammary gland, intraductal papillary adenocarcinoma, lung adenocarcinoma, squamous cell cancer, small-cell cancer, large-cell cancer, glioma which is tumor of neuroepithelial tissue, glioblastoma, neuroblastoma, ependymoma, neuronal tumor, embryonal neuroectodermal tumor, neurilemmoma, neurofibroma, meningioma, chronic lymphocytic leukemia, gastrointestinal lymphoma, alimentary lymphoma, small to medium cell-type lymphoma, cecal cancer, ascending colon cancer, descending colon cancer, transverse colon cancer, sigmoid colon cancer, rectal cancer, epithelial ovarian cancer, germ cell tumor, stromal cell tumor, pan
  • Wilms tumor hepatoblastoma, soft tissue sarcoma, acute leukemia, chronic leukemia, spinal cord tumor, malignant soft tissue tumor, teratoma group tumor, and head and neck cancer including hypopharynx cancer, oropharynx cancer, tongue cancer, epipharynx cancer, oral cancer, lip cancer, sinus cancer, and throat cancer.
  • the recipient subjects are preferably mammals, for example, mammals including primates, pet animals, livestock, and sport animals and are particularly preferably humans, dogs, and cats.
  • the pharmaceutical composition can be formulated by a method generally known to those skilled in the art.
  • the pharmaceutical composition can be used in the form of a parenteral injection of an aseptic solution or suspension with water or any other pharmaceutically acceptable liquid.
  • the pharmaceutical composition may be formulated with the antibody mixed in a unit dosage form required for generally accepted pharmaceutical practice, in appropriate combination with pharmacologically acceptable carriers or media, specifically, sterilized water, physiological saline, a plant oil, an emulsifier, a suspending agent, a surfactant, a stabilizer, a flavoring agent, an excipient, a vehicle, a preservative, a binder, etc.
  • the amount of the active ingredient in such a preparation is determined such that an appropriate dose within the prescribed range can be achieved.
  • An aseptic composition for injection can be formulated according to conventional pharmaceutical practice using a vehicle such as injectable distilled water.
  • aqueous solutions for injection include physiological saline, isotonic solutions containing glucose and other adjuvants, for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride. These solutions may be used in combination with an appropriate solubilizer, for example, an alcohol (specifically, ethanol) or a polyalcohol (e.g., propylene glycol and polyethylene glycol), or a nonionic surfactant, for example, Polysorbate 80TM or HCO-60.
  • an appropriate solubilizer for example, an alcohol (specifically, ethanol) or a polyalcohol (e.g., propylene glycol and polyethylene glycol), or a nonionic surfactant, for example, Polysorbate 80TM or HCO-60.
  • oily solutions examples include sesame oil and soybean oil. These solutions may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizer.
  • the solutions may be further mixed with a buffer (e.g., a phosphate buffer solution and a sodium acetate buffer solution), a soothing agent (e.g., procaine hydrochloride), a stabilizer (e.g., benzyl alcohol and phenol), and an antioxidant.
  • a buffer e.g., a phosphate buffer solution and a sodium acetate buffer solution
  • a soothing agent e.g., procaine hydrochloride
  • a stabilizer e.g., benzyl alcohol and phenol
  • antioxidant e.g., benzyl alcohol and phenol
  • the pharmaceutical composition of the present invention is administered orally or parenterally, preferably parenterally.
  • specific examples of its dosage forms include injections, intranasal administration agents, transpulmonary administration agents, and percutaneous administration agents.
  • the injections include intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection, through which the pharmaceutical composition can be administered systemically or locally.
  • the administration method can be appropriately selected depending on the age, weight, sex, symptoms, etc., of a patient.
  • the dose of a pharmaceutical composition containing the antibody or a polynucleotide encoding the antibody can be selected within a range of, for example, 0.0001 to 10(X) mg/kg of body weight per dose.
  • the dose can be selected within a range of, for example, 0.001 to (X00000 mg/body of a patient, though the dose is not necessarily limited to these numeric values.
  • the dose and the administration method vary depending on the weight, age, sex, symptoms, etc., of a patient, those skilled in the art can appropriately select the dose and the method.
  • the pharmaceutical composition comprising the antibody of the present invention or the fragment thereof can be administered to a subject to treat and/or prevent a cancer, preferably breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor. Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
  • a cancer preferably breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mast
  • the present invention further encompasses a method for treating and/or preventing a cancer, comprising administering the pharmaceutical composition of the present invention in combination with the antitumor agent as exemplified above or a pharmaceutical composition comprising the antitumor agent to a subject.
  • the antibody of the present invention or the fragment thereof may be administered simultaneously with or separately from the antitumor agent to the subject. In the case of separate administration, either of their pharmaceutical compositions may be administered first or later. Their dosing intervals, doses, administration routes, and the number of doses can be appropriately selected by a specialist.
  • the dosage form also includes, for example, a pharmaceutical composition formulated by mixing the antibody of the present invention or the fragment thereof and the antitumor agent in a pharmacologically acceptable carrier (or medium).
  • a pharmaceutical composition formulated by mixing the antibody of the present invention or the fragment thereof and the antitumor agent in a pharmacologically acceptable carrier (or medium).
  • the present invention also provides a combination drug product for the treatment and/or prevention of a cancer, comprising the pharmaceutical composition of the present invention and a pharmaceutical composition comprising the antitumor agent as exemplified above, and a method for treating and/or preventing a cancer, comprising administering the combination drug product.
  • the present invention also provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising the antibody of the present invention or the fragment thereof and the antitumor agent together with a pharmacologically acceptable carrier.
  • the present invention further provides a DNA encoding the antibody of the present invention, a DNA encoding the heavy chain or the light chain of the antibody, and a DNA encoding the heavy chain or light chain variable region of the antibody.
  • DNAs include, in the case of the antibody (a), for example, a DNA encoding a heavy chain variable region, comprising nucleotide sequences encoding the amino acid sequences of SEQ ID NOs: 1, 2, and 3, and a DNA encoding a light chain variable region, comprising nucleotide sequences encoding the amino acid sequences of SEQ ID NOs: 4, 5, and 6.
  • sequences encoding the other regions (i.e., constant regions and framework regions) of the antibody may be sequences derived from a different antibody.
  • the different antibody also includes an antibody derived from a non-human organism, but is preferably one derived from a human from the viewpoint of reduction in adverse reaction.
  • regions encoding the respective framework regions of the heavy chain and the light chain and each constant region should comprise nucleotide sequences encoding the corresponding amino acid sequences derived from a human antibody.
  • DNA encoding the antibody of the present invention include a DNA encoding a heavy chain variable region, comprising a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 8, and a DNA in which a region encoding a light chain variable region comprises a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 15.
  • an example of the nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 8 is the nucleotide sequence of SEQ ID NO: 23.
  • an example of the nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 15 is the nucleotide sequence of SEQ ID NO: 30.
  • regions encoding the respective constant regions of the heavy chain and the light chain should comprise nucleotide sequences encoding the corresponding amino acid sequences derived from a human antibody.
  • RNAs can be obtained by, for example, the method mentioned above or the following method: first, total RNA is prepared from the hybridoma related to the antibody of the present invention using a commercially available RNA extraction kit, and cDNA is synthesized with reverse transcriptase using random primers or the like. Subsequently, the cDNA encoding the antibody is amplified by PCR using, as primers, oligonucleotides having sequences respectively conserved in the respective variable regions of a known mouse antibody heavy chain gene and light chain gene. Sequences encoding constant regions can be obtained by amplifying known sequences by PCR. The nucleotide sequence of the resulting DNA can be determined by a routine method, for example, by integration into a plasmid or a phage for sequencing.
  • the present invention further provides polypeptides and DNAs described in the following (i) to (xv) related to the antibodies (i) to (xiv):
  • a heavy chain CDR polypeptide selected from the group consisting of the amino acid sequences represented by SEQ ID NOs: 1, 2, and 3, and a DNA encoding the polypeptide
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 30, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 10 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 25 or SEQ ID NO: 30, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 30, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 13, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 28, respectively.
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 12, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 27, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 12, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 27, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 13, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 28, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 10 or SEQ ID NO: 14, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 25 or SEQ ID NO: 29, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 11, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 26, respectively.
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 11, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 26, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 9 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 24 or SEQ ID NO: 30, respectively,
  • polypeptide comprising the amino acid sequences of SEQ ID NO: 20 or SEQ ID NO: 21, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 31 or SEQ ID NO: 32, respectively, and
  • (xv) a polypeptide derived from any of the polypeptides described in (i) to (xiv) by comprising the substitution of one or more amino acids in the heavy chain constant region, or a fragment thereof, and a DNA encoding the polypeptide or the fragment thereof.
  • polypeptides and DNAs can be prepared, as described above, using a gene recombination technique.
  • An antibody which comprises a heavy chain variable region comprising complementarity-determining regions of SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising complementarity-determining regions of SEQ ID NOs: 4, 5, and 6, and has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof.
  • An antibody composition comprising an antibody according to (17) and an antibody according to any of (1) to (16) having fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region.
  • a pharmaceutical composition for the treatment and/or prevention of a cancer comprising an antibody or a fragment thereof according to any of (1) to (15), an antibody according to (16) or (17), or an antibody composition according to (18) as an active ingredient.
  • the cancer is breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin
  • a combination drug product for the treatment and/or prevention of a cancer comprising a pharmaceutical composition according to (20) or (21) and a pharmaceutical composition comprising an antitumor agent.
  • a method for treating and/or preventing a cancer comprising administering an antibody or a fragment thereof according to any of (1) to (16), an antibody according to (17), an antibody composition according to (18), a pharmaceutical composition according to (20) or (21), or a combination drug product according to (22) to a subject.
  • Example 3 of WO2010/016526 300 ⁇ g of human CAPRIN-1 protein prepared in Example 3 of WO2010/016526 was mixed with an equal amount of a Freund's complete adjuvant, and this was used as an antigen solution per rabbit. A mixture with a Freund's incomplete adjuvant was used in the second or later immunization.
  • the antigen solution was intraperitoneally administered to each 12-week-old rabbit, and then, administered 8 times every 2 to 3 weeks to complete immunization. Lymphocytes were obtained from the spleen of each rabbit excised 4 days after the final immunization, and mixed with rabbit myeloma cells 240E-W2 at a ratio of 1:2.
  • a PEG solution (heated to 37° C.) prepared by mixing 200 ⁇ L of an RPMI medium containing 10% FBS and 800 ⁇ L of PEG1500 was added thereto, and the mixture was left standing for 5 minutes fuse cells. After centrifugation and removal of the supernatant, the cells were suspended in 300 mL of an RPMI medium containing 10% FBS supplemented with HAT solution (HAT selective medium) at a concentration of 2% and inoculated at 100 ⁇ L/well to 80 96-well plates. Hybridomas generated by the fusion of the spleen cells and the rabbit myeloma cells were obtained by culturing at 37° C. for 7 days under conditions of 5% CO 2 .
  • Hybridomas were selected based on the reactivity of antibodies produced by the prepared hybridomas with the CAPRIN-1 protein.
  • a 1 ⁇ g/mL CAPRIN-1 protein solution was added at 100 ⁇ L/well to the 96-well plates, and the plates were left standing at 4° C. for 18 hours. Each well was washed with PBS-T three times. Then, a 0.5% bovine serum albumin (BSA) solution was added at 400 ⁇ L/well, and the plates were left standing at room temperature for 3 hours. The solution was removed, and the wells were washed with 400 ⁇ L/well of PBS-T three times.
  • BSA bovine serum albumin
  • each culture supernatant of the hybridoma obtained above was added at 100 ⁇ L/well, and the plates were left standing at room temperature for 2 hours. Each well was washed with PBS-T three times. Then, an HRP-labeled anti-rabbit antibody diluted 5000-fold with PBS was added at 100 ⁇ L/well, and the plates were left standing at room temperature for 1 hour. Each well was washed with PBS-T three times. Then, a TMB substrate solution was added at 100 ⁇ L/well, and the plates were left standing for 15 to 30 minutes for chromogenic reaction.
  • the reaction was terminated by the addition of I N sulfuric acid at 100 ⁇ L/well, and the absorbance values were measured at 450 nm and 595 nm using an absorption spectrometer. As a result, a plurality of hybridomas producing antibodies that exhibited a high absorbance values were selected.
  • hybridomas were added at 0.5 cells/well to 96-well plates and cultured. After 1 week, hybridomas that formed single colonies in the wells were observed. The cells in these wells were further cultured, and hybridomas were selected based on the reactivity of antibodies produced by the cloned hybridomas with the CAPRIN-1 protein. As a result of evaluating the reactivity of each antibody with the CAPRIN-1 protein by the same procedure as above, a plurality of hybridoma lines producing rabbit monoclonal antibodies that exhibited reactivity with the CAPRIN-1 protein were obtained.
  • these rabbit monoclonal antibodies that exhibited reactivity with the CAPRIN-1 protein were screened for ones exhibiting reactivity with the surface of human cancer cells on which CAPRIN-1-was expressed.
  • 2 ⁇ 10 5 cells each of a human lung cancer cell line QG56 and a human breast cancer cell line BT-474 (obtained from ATCC) were centrifuged in each 1.5 mL microcentrifuge tube, to which 100 ⁇ L of the culture supernatant of each of the hybridomas was then added. The tube was left standing on ice for 1 hour.
  • variable region-encoding genes as to the rabbit anti-CAPRIN-1 monoclonal antibody obtained above were obtained according to the method described in Example 5 of WO2010/016526, and analyzed for their gene sequences and the amino acid sequences encoded thereby. Specifically. mRNA was extracted from the hybridoma producing the rabbit anti-CAPRIN-1 monoclonal antibody, and the genes of the heavy chain variable (VH) region and the light chain variable (VL) region of this antibody were obtained by RT-PCR using primers specific for rabbit variable region sequences. These genes were inserted to cloning vectors, and their respective nucleotide sequences were determined according to a routine method.
  • the resulting rabbit anti-CAPRIN-1 monoclonal antibody was confirmed to have a heavy chain variable region represented by SEQ ID NO: 20, the heavy chain variable region containing CDR1, CDR2, and CDR3 consisting of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and to have a light chain variable region represented by SEQ ID NO: 21, the light chain variable region containing CDR1.
  • CDR2, and CDR3 consisting of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively.
  • the obtained antibody was reacted with human cancer cells confirmed to express the gene of CAPRIN-1, i.e., breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer
  • CAPRIN-1 i.e., breast cancer cells (BT-474 and MDA-MB-361), color
  • the cells were suspended in 0.5% FBS-PBS( ⁇ ) containing 0.2 ⁇ g/mL (final concentration) propidium iodide, and the fluorescence intensity was measured using FACSCalibur® or FACSVerseTM (Becton, Dickinson and Company).
  • FACSCalibur® or FACSVerseTM Becton, Dickinson and Company.
  • the same procedure as above was carried out for a negative control using a medium for hybridoma culture, and the resultant prepared was used as a sample for a negative control.
  • the fluorescence intensity when using the culture supernatant of the hybridoma producing the rabbit anti-CAPRIN-1 monoclonal antibody was stronger than that when using the negative control. From these results, the rabbit anti-CAPRIN-1 monoclonal antibody was confirmed to react with CAPRIN-1 on the cancer cell membrane surface of human cancer cells.
  • the gene for expressing the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 20 of the rabbit anti-CAPRIN-1 monoclonal antibody confirmed in Example 1 and the gene for expressing the light chain variable region represented by SEQ ID NO: 21 thereof were respectively inserted to a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG and a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1.
  • the prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant containing a human-rabbit chimeric anti-CAPRIN-1 antibody (human-rabbit chimeric antibody) was obtained.
  • the obtained culture supernatant containing the chimerized antibody was purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.).
  • the buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare the chimeric antibody.
  • a nucleotide sequence was designed so as to be able to express the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 7 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively. This was inserted to a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1.
  • nucleotide sequence was designed so as to be able to express the amino acid sequence of a light chain variable region represented by SEQ ID NO: 11 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively. This was inserted to a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1.
  • a culture supernatant containing humanized antibody #1 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 8 in which heavy chain variable region CDR1.
  • CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: I, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the amino acid sequence of the light chain variable region represented by SEQ ID NO: 11, was obtained.
  • culture supernatants containing the following humanized antibodies #2 to 10 were further obtained:
  • humanized antibody #2 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 12 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #3 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 13 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #4 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 12;
  • humanized antibody #5 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 13;
  • humanized antibody #6 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #7 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15;
  • humanized antibody #8 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 9 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the light chain full-length amino acid sequence represented by SEQ ID NO: 15.
  • humanized antibody #9 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 10 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the light chain full-length amino acid sequence represented by SEQ ID NO: 14 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively; and
  • humanized antibody #10 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 10 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15:
  • the obtained culture supernatants containing the humanized antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare the humanized antibodies.
  • Example 2 the specific reactivity of the human-rabbit chimeric antibody prepared in Example 2 and the humanized antibodies #0 to #10 prepared in Example 3 with the CAPRIN-1 protein was confirmed by ELISA according to a routine method. Specifically, at first, a PBS solution containing 5 ⁇ g/mL CAPRIN-1 protein was added at 100 ⁇ L/well to 96-well plates, and the plates were left standing at 4° C. for 18 hours. Each well was washed with PBS-T. Then, a blocking solution consisting of a PBS solution containing 5% skimmed milk was added at 400 ⁇ L/well, and the plates were left standing at room temperature for 3 hours.
  • an HRP-labeled anti-human IgG antibody diluted 3000-fold with PBS containing 0.2% skimmed milk was added at 50 ⁇ L/well, and the plates were left standing at room temperature for 1 hour. Each well was washed with PBS-T three times. Then, a TMB substrate solution (manufactured by Thermo Fischer Scientific, Inc.) was added at 100 ⁇ l/well, and the plates were left standing for 1 to 30 minutes for chromogenic reaction. After the color development, the reaction was terminated by the addition of 1 N sulfuric acid at 100 ⁇ L/well, and the absorbance values were measured at 450 nm and 630 nm using an absorption spectrometer.
  • the purified human-rabbit chimeric antibody and humanized antibodies #0 to #10 were each reacted with human cancer cells confirmed to express the gene of CAPRIN-1, i.e., breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gall
  • each cancer cell line was collected into each 1.5 mL microcentrifuge tube, and each of the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 was added at 50 ⁇ g/mL (final concentration) to each tube and reacted at 4° C. for 1 hour.
  • an Alexa 488-labeled goat anti-human IgG (H+L) antibody (manufactured by Life Technologies Corp.) diluted 100-fold with 0.5% FBS-PBS( ⁇ ) was added thereto, and the tube was left standing at 4° C. for 60 minutes.
  • the cells were suspended in 0.5% FBS-PBS( ⁇ ) containing 0.2 ⁇ g/mL (final concentration) propidium iodide, and the fluorescence intensity was measured using FACSCaliburTM or FACSVerseTM (Becton, Dickinson and Company).
  • FACSCaliburTM or FACSVerseTM Becton, Dickinson and Company.
  • the same procedure as above was carried out using a medium for hybridoma culture, and the resultant prepared was used for a negative control.
  • the fluorescence intensity from the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 was stronger than that in the case of using the negative control. From these results, the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 were confirmed to react with the CAPRIN-1 protein expressed on the membrane surface of human cancer cells.
  • Example 2 human-rabbit chimeric antibody prepared in Example 2 and the humanized antibodies #0 to #10 prepared in Example 3 were evaluated for their antitumor effects on various human cancer cells on the basis of ADCC activity.
  • anti-CAPRIN-1 antibodies were used as comparative antibodies for the human-rabbit chimeric antibody and the humanized antibodies #0 to #10:
  • an antibody described in WO2011/096519 i.e., comparative antibody 20 having a heavy chain variable region of SEQ ID NO: 42 and a light chain variable region of SEQ ID NO: 46 in this literature;
  • an antibody described in WO2013/018892 i.e., comparative antibody 34 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2013/018891 i.e., comparative antibody 35 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2013/018889 i.e., comparative antibody 36 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • comparative antibody 39 having a heavy chain variable region of SEQ ID NO: 52 and a light chain variable region of SEQ ID NO: 54 in this literature
  • comparative antibody 40 having a heavy chain variable region of SEQ ID NO: 21 and a light chain variable region of SEQ ID NO: 23 therein
  • comparative antibody 41 having a heavy chain variable region of SEQ ID NO: 25 and a light chain variable region of SEQ ID NO: 23 therein
  • comparative antibody 42 having a heavy chain variable region of SEQ ID NO: 16 and a light chain variable region of SEQ ID NO: 18 therein
  • comparative antibody 43 having a heavy chain variable region of SEQ ID NO: 29 and a light chain variable region of SEQ ID NO: 33 therein
  • comparative antibody 44 having a heavy chain variable region of SEQ ID NO: 39 and a light chain variable region of SEQ ID NO: 43 therein
  • comparative antibody 45 having a heavy chain variable region of SEQ ID NO: 49 and a light chain variable region of SEQ ID NO: 49 and a light chain
  • the aforementioned compared antibodies were each prepared as follows: the gene for expressing the amino acid sequence of the heavy chain variable region and the gene for expressing the light chain variable region were respectively inserted to a vector pcDNA4/myc-His for expression in mammalian cells (manufactured by Life Technologies Corp.) having a gene insert of the heavy chain constant region of human IgG1 and a vector pcDNA3.1/myc-His for expression in mammalian cells (manufactured by Life Technologies Corp.) having a gene insert of the light chain constant region of human IgG1; the prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method; the obtained human-chimerized or humanized antibody was purified using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.); the buffer was replaced with PBS( ⁇ ); and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Mer
  • a well to which an isotype control antibody was added, a well to which no antibody was added, and a well to which an antibody reacting with the CAPRIN-1 protein but exhibiting no reactivity with the surface of human cancer cells, on which CAPRIN-1 was expressed, was added were prepared as negative controls.
  • Each antibody was added at 5 ⁇ g/mL (final concentration) to V-bottomed 96-well plates.
  • Human NK cells separated from human peripheral blood mononuclear cells by using a routine method were used as effector cells.
  • the human peripheral blood mononuclear cells were separated using a specific gravity separation solution Histopaque for peripheral blood mononuclear cell separation (Sigma-Aldrich Corp.), and reacted with antibodies (anti-human CD3 antibody, anti-human CD20 antibody, anti-human CD19 antibody, anti-human CD11c antibody, anti-HLA-DR antibody (BD Pharmingen)) labeled with an FITC fluorescent dye.
  • a cell population containing NK cells that were not stained with these antibodies was separated using a cell sorter (FACS Vantage SE (Becton, Dickinson and Company)).
  • a cell population separated using a human NK cell separation kit (manufactured by Miltenyi Biotec K.K.) was used.
  • a culture supernatant containing chromium 51 released into the culture supernatant from damaged cancer cells was recovered from each well, added to LumaPlate-96 (manufactured by PerkinElmer, Inc.) with the bottom of each well coated with a solid scintillator, and dried.
  • the amount of chromium 51 released into the culture supernatant from damaged cancer cells was measured to calculate the antitumor effects of the anti-CAPRIN-1 antibodies on the cancer cells.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 54% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 50% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 46% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 25% or lower activity, and all of the negative control groups exhibited 10% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 52% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 25% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 43% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 35% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 3% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 46% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 42% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 38% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 22% or lower activity
  • all of the negative control groups exhibited 10%0 or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 45% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 38% or higher activity
  • the antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 34% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 8% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 52% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 49% or higher antitumor effect
  • the humanized antibody #3, the antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 38% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 12% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 24% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 2% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 28% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 25% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 21% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 12% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 31% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 27% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 37% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 26% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 36% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 29% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 24% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 36% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 45% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 28% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity
  • all of the negative control groups exhibited 7% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 20% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 18% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 20% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 18% or higher activity
  • the antibody #9, the antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 25% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 30% or higher antitumor effect
  • the humanized antibody #3, the antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 25% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 20% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 25% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 21% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 8% or lower activity
  • all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 50% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 46% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 8% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 48% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 31% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 40% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 36% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 39% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 31% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the antibody #2, and the humanized antibody #5 exhibited 30% or higher activity
  • the humanized antibody #9, the antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 27% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 10%0 or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 37% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 25% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 11% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 42% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 35% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 50% or higher antitumor effect
  • the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity
  • the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the antibody #8 exhibited 35% or higher activity.
  • all of the comparative antibodies 1 to 48 exhibited 20% or lower activity
  • all of the negative control groups exhibited 8% or lower activity.
  • humanized antibodies #0 to #10 and the human-rabbit chimeric antibody exhibit a significantly stronger antitumor effect on breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer than that of the comparative antibodies.
  • the humanized antibodies #0 to #10 and the human-rabbit chimeric antibody exhibited significantly stronger antitumor activity against breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer as described above than that of all of the antibodies against CAPRIN-1 described in Examples of WO2010/016526, WO2011/096517, WO2011/096528, WO2011/096519.
  • WO2011/096533 WO2011/096534.
  • WO2011/096535 WO2013/018886.
  • WO20131018894 WO2013/018892, WO2013/018891, WO2013/018889, WO2013/018883, WO2013/125636, WO2013/125654, WO2013/125630, WO2013/125640, WO2013/147169, and WO2013/147176.
  • the antitumor effect was shown as cytotoxic activity against the cancer cell lines that was determined by mixing each antibody against CAPRIN-1, effector cells, and chromium 51-incorporated target cells, culturing the cells for 4 hours, and measuring the amount of chromium 51 released into the medium after the cultivation, followed by calculating according to the following formula*:
  • Anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 33 in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 was substituted (hereinafter, referred to as engineered I-type anti-CAPRIN-1 antibodies) were prepared.
  • a DNA encoding the amino acid sequence of a heavy chain having the heavy chain constant region mentioned above and a heavy chain variable region represented by SEQ ID NO: 7 was synthesized, and this was inserted to a vector for expression in mammalian cells according to a routine method.
  • a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was inserted to a vector for expression in mammalian cells having an insert of a gene encoding the light chain constant region of human IgG to prepare a recombinant expression vector.
  • the prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant of engineered I-type anti-CAPRIN-1 antibody #0 of the humanized antibody #0 was obtained.
  • Culture supernatants containing engineered I-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3 were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively.
  • the obtained culture supernatants containing the engineered I-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare the engineered I-type anti-CAPRIN-1 antibodies.
  • anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 34 in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 was substituted (hereinafter, referred to as engineered II-type anti-CAPRIN-1 antibodies), were prepared.
  • a DNA encoding the amino acid sequence of a heavy chain having the heavy chain constant region mentioned above and a heavy chain variable region represented by SEQ ID NO: 7 was synthesized, and this was inserted to a vector for expression in mammalian cells according to a routine method.
  • a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was inserted to a vector for expression in mammalian cells having an insert of a gene encoding the light chain constant region of human IgG1 to prepare a recombinant expression vector.
  • These two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant of engineered II-type anti-CAPRIN-1 antibody #0 was obtained.
  • the obtained culture supernatants containing the engineered II-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare the engineered II-type anti-CAPRIN-1 antibodies.
  • anti-CAPRIN-1 antibodies having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 (hereinafter, referred to as engineered III-type anti-CAPRIN-1 antibodies) were obtained by the following method: a neomycin resistance gene-containing mammalian cell expression vector harboring a gene of GDP-6-deoxy-D-lyxo-4-hexulose reductase (RMD), which is an enzyme that does not catalyze the reaction of converting GDP-6-deoxy-D-lyxo-4-hexulose to GDP-L-fucose, was transferred to mammalian cell line CHO cells according to a routine method using a transfection reagent FreeStyleTM MAX Reagent (Life Technologies Corp.
  • the transfected CHO cells were cultured in a medium containing G-418 to prepare a stable pool of CHO cells expressing RMD. From this stable pool, 7 CHO cells constitutively expressing RMD were cloned by the limiting dilution method. The RMD gene expression level in each of the cloned 7 RMD-CHO cells was evaluated three times every other week by quantitative PCR to select CHO cells constitutively and stably expressing the RMD gene (RMD-CHO cells).
  • the gene encoding the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the gene encoding the amino acids of the light chain variable region represented by SEQ ID NO: 11 were respectively transferred through a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1 and a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1 to the RMD-CHO cells constitutively expressing RMD according to a routine method, and a culture supernatant containing engineered III-type anti-CAPRIN-1 antibody #0 was obtained.
  • the obtained culture supernatants containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.).
  • the buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare an antibody composition containing the engineered III-type anti-CAPRIN-1 antibody #0. Similarly, antibody purified products containing engineered III-type anti-CAPRIN-1 antibodies #1 to #10 were obtained.
  • the proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of these purified antibody compositions was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • the gene encoding the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the gene encoding the amino acids of the light chain variable region represented by SEQ ID NO: 11 were respectively transferred through a hygromycin resistance gene-containing vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1 and a hygromycin resistance gene-containing vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1 to the RMD-CHO cells according to a routine method, and the resulting cells were cultured in a medium containing hygromycin B to prepare a stable pool expressing engineered III-type anti-CAPRIN-1 antibody #0.
  • anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 34 in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 described in Example 3 was substituted, and having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region of the antibody (hereinafter, referred to as engineered N-type anti-CAPRIN-1 antibodies) were prepared.
  • a DNA encoding the amino acid sequence of a heavy chain having the variant heavy chain constant region prepared in Example 6-1 and the heavy chain variable region of human IgG1 represented by SEQ ID NO: 7 was synthesized and inserted to a vector for expression in mammalian cells according to a routine method, while a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was synthesized and inserted to a vector for expression in mammalian cells having an insert of a gene encoding the amino acids of the light chain constant region of human IgG1, and the resulting vectors were transferred to the RMD-CHO cells constitutively expressing GDP-6-deoxy-D-lyxo-4-hexulose reductase (RMD) prepared in Example 6-2.
  • RMD RMD-CHO cells constitutively expressing GDP-6-deoxy-D-lyxo-4-hexulose reductase
  • a culture supernatant of engineered N-type anti-CAPRIN-1 antibody #0 of the humanized antibody #0 was obtained.
  • Culture supernatants containing engineered N-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively, described in Example 3.
  • the obtained culture supernatants containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.).
  • the buffer was replaced with PBS( ⁇ ), and the resultant was filtered through a 0.22 ⁇ m filter (manufactured by Merck Millipore Corp.) to prepare an antibody composition containing the engineered IV-type anti-CAPRIN-1 antibody #0. Similarly, antibody purified products containing engineered IV-type anti-CAPRIN-1 antibodies #1 to #10 were obtained.
  • the proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of these purified antibody compositions was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • a DNA encoding the amino acid sequence of a heavy chain having the modified heavy chain constant region prepared in Example 6-1 and the heavy chain variable region of human IgG1 represented by SEQ ID NO: 7 was synthesized and inserted to a hygromycin resistance gene-containing vector for expression in mammalian cells according to a routine method, while a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was synthesized and inserted to a hygromycin resistance gene-containing vector for expression in mammalian cells having an insert of a gene encoding the amino acids of the light chain constant region of human IgG1, and the resulting vectors were transferred to the cells.
  • the cells were cultured in a medium containing hygromycin B to prepare a stable pool expressing engineered IV-type anti-CAPRIN-1 antibody #0. From this stable pool, cells constitutively and stably expressing the engineered IV-type anti-CAPRIN-1 antibody #0 were prepared by the limiting dilution method. Cells constitutively and stably expressing engineered IV-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 were further prepared in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3, respectively.
  • the proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of the purified antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 produced by the respective cells was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • engineered-type anti-CAPRIN-1 antibodies #0 to #10 The specific reactivity of the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 (hereinafter, referred to as engineered-type anti-CAPRIN-1 antibodies) prepared in Examples 6-1 to 6-3 with the CAPRIN-1 protein was confirmed in the same way as in Example 4.
  • the engineered-type anti-CAPRIN-1 antibodies (the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10) prepared in Examples 6-1 to 6-3 were evaluated for their antitumor effects on various human cancer cells on the basis of ADCC activity in the same way as in Example 5.
  • a well to which an isotype control antibody was added, a well to which no antibody was added, and a well to which an antibody reacting with the CAPRIN-1 protein but exhibiting no reactivity with the surface of human cancer cells, on which CAPRIN-1 was expressed, was added were prepared as negative controls.
  • Human NK cells separated from human peripheral blood mononuclear cells by using a routine method were used as effector cells.
  • the human NK cells added at 0.4 to 2.0 ⁇ 10 5 cells/well to the V-bottomed 96-well plates to which each of the antibodies and the target cells were added and reacted were prepared and reacted at 37° C. for 4 hours under conditions of 5% CO 2 .
  • 50 ⁇ L of a culture supernatant containing chromium 51 released from damaged cancer cells was recovered from each well.
  • the amount of chromium 51 released into the culture supernatant from damaged cancer cells was measured in the same way as in Example 5 to calculate the antitumor effects of the anti-CAPRIN-1 antibodies on the cancer cells.
  • the engineered-type anti-CAPRIN-1 antibodies i.e., the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 each exhibited a stronger antitumor effect than that of the negative controls.
  • the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 exhibited the same level of the antitumor effect as that exhibited by the corresponding unengineered antibodies (humanized antibodies #0 to #10) used as comparative antibodies were approximately 1/13 to 1/20 concentrations of the unengineered antibody concentrations, respectively.
  • the concentrations were approximately 1/150 of the unengineered antibody concentrations, respectively.
  • the breast cancer cells MDA-MB-361
  • the colorectal cancer cells HT-29
  • the lung cancer cells QG56
  • the stomach cancer cells NCI-N87
  • the uterine cancer cells HEC-1-A
  • the prostate cancer cells 22Rv1
  • the pancreatic cancer cells Panc10.5
  • the liver cancer cells Hep3B
  • the ovary cancer cells SKOV3
  • the kidney cancer cells Caki-2
  • the brain tumor cells U-87MG
  • the bladder cancer cells T24 and HT-1376
  • the esophagus cancer cells OE33
  • the leukemia cells OCI-AML5
  • the lymphoma cells Ramos
  • the gallbladder cancer cells TGBC14TKB
  • the fibrosarcoma cells HT-1080
  • the melanoma cells G-361
  • the adrenal cortex cancer cells A-673
  • the Ewing's tumor cells RD-ES
  • the antibody of the present invention is useful for the treatment and/or prevention of a cancer.

Abstract

It is intended to provide a CAPRIN-1-targeting antibody superior in antitumor activity to conventional antibodies, and use thereof as a therapeutic and/or preventive agent for a cancer. The present invention provides an antibody targeting a CAPRIN-1 polypeptide specifically expressed on the surface of cancer cells, and use of the antibody as a therapeutic and/or preventive agent for a cancer. The present invention provides an antibody which comprises a heavy chain variable region comprising complementarity-determining regions of SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising complementarity-determining regions of SEQ ID NOs: 4, 5, and 6 and has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof, and a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising this antibody or fragment as an active ingredient.

Description

    TECHNICAL FIELD
  • The present invention relates to an antibody against CAPRIN-1 or a fragment thereof, and novel pharmaceutical use thereof as a therapeutic and/or preventive agent for a cancer, etc.
  • BACKGROUND ART
  • Cancers are diseases that account for the leading cause of death. The current treatment thereof consists principally of surgical therapy, which may be combined with radiation therapy and/or chemotherapy. In spite of the development of novel surgical techniques or the discovery of novel anticancer agents in recent years, the existing treatment of cancers has an insufficiently improved outcome, except for some cancers. With advances of molecular biology or cancer immunology, antibodies that specifically react with cancers, cancer antigens that are recognized by cytotoxic T cells, genes encoding such cancer antigens, and the like have been identified in recent years, raising expectations on specific cancer therapy targeting the cancer antigens.
  • Cytoplasmic-activation and proliferation-associated protein 1 (CAPRIN-1) has been known as an intracellular protein that is expressed upon activation or cell division of resting normal cells and forms cytoplasmic stress granules with intracellular RNAs to participate in the regulation of transport and translation of mRNAs. This protein has been found to be specifically expressed on the surface of cancer cells and is under study as a target of antibody drugs for cancer treatment (Patent Literatures 1 to 19).
  • CITATION LIST Patent Literature
    • Patent Literature 1: WO2010/016526
    • Patent Literature 2: WO2011/096517
    • Patent Literature 3: WO2011/096528
    • Patent Literature 4: WO2011/096519
    • Patent Literature 5: WO2011/096533
    • Patent Literature 6: WO2011/096534
    • Patent Literature 7: WO2011/096535
    • Patent Literature 8: WO2013/018886
    • Patent Literature 9: WO2013/018894
    • Patent Literature 10: WO2013/018892
    • Patent Literature 11: WO2013/018891
    • Patent Literature 12: WO2013/018889
    • Patent Literature 13: WO2013/018883
    • Patent Literature 14: WO2013/125636
    • Patent Literature 15: WO2013/125654
    • Patent Literature 16: WO2013/125630
    • Patent Literature 17: WO2013/125640
    • Patent Literature 18: WO2013/147169
    • Patent Literature 19: WO2013/147176
    SUMMARY OF INVENTION Technical Problem
  • An object of the present invention is to produce an antibody that targets CAPRIN-1 specifically expressed on the surface of cancer cells and is superior in antitumor activity to conventional antibodies, and to provide use thereof as a therapeutic and/or preventive agent for a cancer.
  • Solution to Problem
  • Features of the present invention are as follows:
  • The present invention provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising an antibody which comprises a heavy chain variable region comprising SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising SEQ ID NOs: 4, 5, and 6 and has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof as an active ingredient.
  • In an embodiment thereof, the cancer is breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
  • In another embodiment, the antibody is a human antibody, a humanized antibody, a chimeric antibody, a single-chain antibody, or a multispecific antibody (e.g., a bispecific antibody).
  • The present specification encompasses the contents described in the specification and/or drawings of Japanese Patent Application No. 2013-166164 on which the priority of the present application is based.
  • Advantageous Effects of Invention
  • The antibody against CAPRIN-1 according to the present invention damages cancer cells. Thus, the antibody against CAPRIN-1 according to the present invention is useful in the treatment and/or prevention of a cancer.
  • DESCRIPTION OF EMBODIMENTS
  • The antibody against a polypeptide of CAPRIN-1 used in the present invention can be evaluated for its antitumor activity, as mentioned later, by examining ex vivo whether or not to exhibit immunocyte-mediated cytotoxic activity against tumor cells expressing the polypeptide or by examining in vivo the inhibition of tumor growth in a cancer-bearing animal.
  • The antibody against CAPRIN-1 according to the present invention may be a monoclonal antibody or a polyclonal antibody and is preferably a monoclonal antibody. The antibody against CAPRIN-1 according to the present invention may be any type of antibody that can exert antitumor activity and includes, for example, recombinant antibodies (e.g., synthetic antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized antibodies, chimeric antibodies, and single-chain antibodies (scFv)), human antibodies, and their antibody fragments (e.g., Fab, F(ab′)2, and Fv). These antibodies and fragments thereof can be prepared by methods generally known to those skilled in the art. When a subject is a human, a human antibody or a humanized antibody is desirable for circumventing or suppressing rejection.
  • The phrase “specifically binding to a CAPRIN-1 protein” means that the antibody specifically binds to the CAPRIN-1 protein without substantially binding to other proteins.
  • The subject according to the present invention, whose cancer is to be treated and/or prevented is a mammal such as a human, a pet animal, livestock, or a sport animal, and a preferred subject is a human.
  • Hereinafter, the preparation of the antigen, the preparation of the antibody, and the pharmaceutical composition according to the present invention will be described.
  • <Preparation of Antigen for Antibody Preparation>
  • A proteins or a fragment thereof used as a sensitizing antigen for obtaining the antibody against CAPRIN-1 according to the present invention is not limited by animal species serving as the origin thereof, including humans, dogs, cats, cattle, horses, mice, rats, and chickens. However, it is preferred to select the sensitizing antigen in view of compatibility with parent cells for use in cell fusion. In general, a mammal-derived protein is preferred. Particularly, a human-derived protein is preferred. For example, when CAPRIN-1 is human CAPRIN-1, the human CAPRIN-1 protein, a partial peptide thereof, cells expressing human CAPRIN-1, or the like can be used.
  • The nucleotide sequences and amino acid sequences of human CAPRIN-1 and homologs thereof can be obtained, for example, by making an access to GenBank (NCBI, USA) and using an algorithm such as BLAST or FASTA (Karlin and Altschul, Proc. Natl. Acad. Sci. USA, 90: 5873-5877, 1993, and Altschul et al., Nucleic Acids Res. 25: 3389-3402, 1997).
  • In the present invention, with reference to the nucleotide sequence (SEQ ID NO: 16 or 18) or amino acid sequence (SEQ ID NO: 17 or 19) of human CAPRIN-1, the target CAPRIN-1 is a nucleic acid or a protein consisting of a sequence having 70% to 100%, preferably 80% to 100%, more preferably 90% to 100%, further preferably 95% to 100%, for example, 97% to 100%, 98% to 100%, 99% to 100%, or 99.5% to 100% sequence identity to the nucleotide sequence or amino acid sequence of the ORF or mature portion of the reference (the amino acid sequences of SEQ ID NO: 17 and SEQ ID NO: 19 compared with each other differ in amino acid residues at and following position 690). In this context, the term “% sequence identity” means a percentage (%) of the number of identical amino acids (or bases) to the total number (including the number of gaps) of amino acids (or bases) when two sequences are aligned such that the maximum degree of similarity or identity can be achieved with or without introduced gaps.
  • The fragment of the CAPRIN-1 protein has a length ranging from the amino acid length of an epitope (antigenic determinant), which is the smallest unit recognized by the antibody, to less than the full length of the protein. The epitope refers to a polypeptide fragment having antigenicity or immunogenicity in mammals, preferably humans. Its smallest unit consists of approximately 7 to 12 amino acids, for example, 8 to 11 amino acids.
  • A polypeptide fragment comprising the aforementioned human CAPRIN-1 protein or a partial peptide thereof can be synthesized according to a chemical synthesis method, for example, an Fmoc (fluorenylmethyloxycarbonyl) or tBoc (t-butyloxycarbonyl) method (Seikagaku Jikken Koza 1 (Biochemical Experimentation Course 1 in English), the Japanese Biochemical Society ed., Protein Chemistry IV. Chemical Modification and Peptide Synthesis, Tokyo Kagaku Dojin Co., Ltd. (Japan), 1981). Also, the human CAPRIN-1 protein or the polypeptide fragment can be synthesized by a routine method using various commercially available peptide synthesizers. Alternatively, a polynucleotide encoding the polypeptide is prepared using a genetic engineering approach known in the art (Sambrook et al., Molecular Cloning, the 2nd edition. Current Protocols in Molecular Biology (1989), Cold Spring Harbor Laboratory Press: Ausubel et al., Short Protocols in Molecular Biology, the 3rd edition, A compendium of Methods from Current Protocols in Molecular Biology (1995). John Wiley & Sons: etc.), and this polynucleotide is incorporated into expression vectors, which are then transferred to host cells so that the polypeptide is produced in the host cells. In this way, the human CAPRIN-1 protein of interest or the polypeptide fragment thereof can be obtained.
  • The polynucleotide encoding the polypeptide can be readily prepared by a genetic engineering approach known in the art or a routine method using a commercially available nucleic acid synthesizer. For example, a DNA comprising the nucleotide sequence of human CAPRIN-1 gene can be prepared by PCR using a human chromosomal DNA or cDNA library as a template and a pair of primers designed so as to be able to amplify the nucleotide sequence. Reaction conditions for this PCR can be appropriately determined. Examples of the conditions can include, but are not limited to, 30 cycles each involving reaction steps consisting of 94° C. for 30 seconds (denaturation), 55° C. for 30 seconds to 1 minute (annealing), and 72° C. for 2 minutes (elongation) using thermostable DNA polymerase (e.g., Taq polymerase or Pfu polymerase) and a Mg2+-containing PCR buffer, followed by reaction at 72° C. for 7 minutes. The PCR approach, conditions, etc., are described in, for example, Ausubel et al., Short Protocols in Molecular Biology, the 3rd edition, A Compendium of Methods from Current Protocols in Molecular Biology (1995), John Wiley & Sons (particularly, Chapter 15).
  • Also, appropriate probes or primers can be prepared on the basis of information on the nucleotide sequence of the CAPRIN-1 gene and the amino acid sequence of the CAPRIN-1 protein, and used in the screening of, for example, a human cDNA library, to isolate the desired DNA. It is preferred that the cDNA library should be produced from cells, organs, or tissues expressing the CAPRIN-1 protein. Examples of such cells or tissues include cells or tissues derived from the testis as well as from cancers or tumors such as leukemia, breast cancer, lymphoma, brain tumor, lung cancer, pancreatic cancer, colorectal cancer, kidney cancer, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, sarcoma, mastocytoma, liver cancer, gallbladder cancer, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, and head and neck cancer. These procedures, including the preparation of probes or primers, the construction of a cDNA library, the screening of the cDNA library, and the cloning of the gene of interest, are known to those skilled in the art and can be carried out according to methods described in, for example. Sambrook et al., Molecular Cloning, the 2nd edition, Current Protocols in Molecular Biology (1989), and Ausubel et al. (ibid.). The DNA encoding the human CAPRIN-1 protein or the partial peptide thereof can be obtained from the DNA thus obtained.
  • The host cells to receive the expression vectors may be any cell capable of expressing the polypeptide. Examples of prokaryotic cells include, but are not limited to, E. coli. Examples of eukaryotic cells include, but are not limited to: mammalian cells such as monkey kidney cells COS 1 and Chinese hamster ovary cells CHO; a human embryonic kidney cell line HEK293; a mouse embryonic skin cell line NIH3T3; yeast cells such as budding yeast and fission yeast cells; silkworm cells; and Xenopus egg cells.
  • In the case of using prokaryotic cells as the host cells, expression vectors having an origin that permits replication in the prokaryotic cells, a promoter, a ribosomal binding site, a multicloning site, a terminator, a drug resistance gene, an auxotrophic complementary gene, etc., are used as the expression vectors. Examples of expression vectors for E. coli can include pUC series, pBluescript II, pET expression systems, and pGEX expression systems. The DNA encoding the polypeptide is incorporated into such expression vectors, and prokaryotic host cells can be transformed with these vectors, followed by the culture of the obtained transformants so that the polypeptide encoded by the DNA is expressed in the prokaryotic host cells. In this respect, the polypeptide may be expressed as a fusion protein with another protein.
  • In the case of using eukaryotic cells as the host cells, expression vectors for eukaryotic cells having a promoter, a splicing region, a poly(A) addition site, etc., are used as the expression vectors. Examples of such expression vectors can include pKA1, pCDM8, pSVK3, pMSG, pSVL, pBK-CMV, pBK-RSV, EBV, pRS, pcDNA3, and pYES2 vectors. In the same way as above, the DNA encoding the polypeptide is incorporated into such expression vectors, and eukaryotic host cells can be transformed with these vectors, followed by the culture of the obtained transformants so that the polypeptide encoded by the DNA is expressed in the eukaryotic host cells. In the case of using expression vectors such as pIND/V5-His, pFLAG-CMV-2, pEGFP-N1, or pEGFP-C1, the polypeptide can be expressed as various fusion proteins tagged with His tag (e.g., (His)6 to (His)10), FLAG tag, myc tag. HA tag, GFP, or the like.
  • The transfer of the expression vectors to the host cells can employ a well-known method such as electroporation, a calcium phosphate method, a liposome method, a DEAE dextran method, microinjection, viral infection, lipofection, or binding with cell-penetrating peptides.
  • Separation treatments known in the art can be carried out in combination for isolating and purifying the polypeptide of interest from the host cells. Examples thereof include, but are not limited to, treatment with a denaturant (e.g., urea) or a surfactant, ultrasonication, enzymatic digestion, salting-out, solvent fractionation and precipitation, dialysis, centrifugation, ultrafiltration, gel filtration, SDS-PAGE, isoelectric focusing electrophoresis, ion-exchange chromatography, hydrophobic chromatography, affinity chromatography, and reverse-phase chromatography.
  • The antigen thus prepared can be used as a sensitizing antigen as mentioned later for producing the antibody according to the present invention.
  • <Structure of Antibody>
  • Antibodies (immunoglobulins) are usually heteromultimeric glycoproteins each comprising at least two heavy chains and two light chains. The immunoglobulins, except for IgM, are heterotetrameric glycoproteins of approximately 150 kDa each composed of two identical light (L) chains and two identical heavy (H) chains. Typically, each light chain is connected to a heavy chain via a single covalent disulfide bond, though the number of disulfide bonds varies among heavy chains of different immunoglobulin isotypes. Each heavy chain and light chain also have an intrachain disulfide bond. Each heavy chain has a variable domain (VH region) at one end, followed by a series of constant regions. Each light chain has a variable domain (VL region) at one end and has a single constant region at the other end. The light chain constant region is aligned with the first heavy chain constant region, while the light chain variable domain is aligned with the heavy chain variable domain. Particular regions called complementarity-determining regions (CDRs) in the antibody variable domains exhibit specific variability and impart binding specificity to the antibody. Moietys relatively conserved in the variable regions are called framework regions (FRs). The complete heavy chain and light chain variable domains each has a structure in which four framework regions are connected via three complementarity-determining regions, i.e., a structure in which FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4 are connected in this order from the N-terminus. These three complementarity-determining regions in the heavy chain are called CDRH1, CDRH2, and CDRH3 in this order from the N-terminus thereof. Likewise, the complementarity-determining regions in the light chain are called CDRL1, CDRL2, and CDRL3. CDRH3 is most important for the binding specificity of the antibody for its antigen. In addition, CDRs in each chain are kept close to each other by the framework regions and contribute to the formation of an antigen-binding site in the antibody, together with the complementarity-determining regions derived from the other chain. The constant regions do not directly contribute to antibody-antigen binding, but exhibit various effector functions, for example, involvement in antibody-dependent cellular cytotoxicity (ADCC), phagocytosis (ADCP) mediated by binding to an Fcγ receptor, half-life/clearance rates mediated by a neonatal Fc receptor (FcRn), and complement-dependent cytotoxicity (CDC) mediated by a Clq component in the complement cascade.
  • <Preparation of Antibody>
  • The anti-CAPRIN-1 antibody according to the present invention means an antibody having immunological reactivity with a full-length CAPRIN-1 protein or a fragment thereof.
  • In this context, the “immunological reactivity” means the property of the antibody binding to the CAPRIN-1 antigen (a full-length CAPRIN-1 protein or a partial polypeptide thereof) in vivo. Via such binding to CAPRIN-1, the antibody of the present invention exerts the function of damaging (e.g., killing, suppressing, or regressing) tumor cells. The antibody of the present invention can damage tumor, for example, breast cancer, kidney cancer, pancreatic cancer, colorectal cancer (e.g., colon cancer), lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer. Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer through binding to the CAPRIN-1 protein.
  • The antibody of the present invention is not particularly limited, preferably, as long as the antibody is a monoclonal antibody. The antibody of the present invention includes synthetic antibodies, multispecific antibodies (e.g., diabodies and triabodies), human antibodies, humanized antibodies, chimeric antibodies, single-chain antibodies, antibody fragments (e.g., Fab, F(ab′)2, and Fv), and the like. Also, the antibody is an immunoglobulin molecule of any class, for example, IgG, IgE, IgM, IgA, IgD, or IgY, or of any subclass, for example, IgG1, IgG2, IgG3, IgG4, IgA1, or IgA2.
  • The antibody may be further modified by deglycosylation, acetylation, formylation, amidation, phosphorylation, PEGylation, or the like, in addition to glycosylation.
  • Hereinafter, preparation examples of various monoclonal antibodies will be given.
  • For example, a breast cancer cell line SK-BR-3 expressing CAPRIN-1 is administered to a mouse for immunization. The spleen is extracted from this mouse. After separation of spleen cells, the cells are fused with mouse myeloma cells. Clones producing antibodies having a cancer cell growth inhibitory effect are selected from among the obtained fusion cells (hybridomas). The hybridomas producing monoclonal antibodies having a cancer cell growth inhibitory effect are isolated, and these hybridomas are cultured. The antibody of the present invention can be prepared by purification from the culture supernatant according to a general affinity purification method.
  • The monoclonal antibody-producing hybridomas may be prepared, for example, as follows: first, animals are immunized with the sensitizing antigen according to a method known in the art. This immunization method generally involves intraperitoneally or subcutaneously injecting the sensitizing antigen to mammals. Specifically, the sensitizing antigen diluted with or suspended in PBS (phosphate-buffered saline), physiological saline, or the like into an appropriate amount is mixed, if desired, with an appropriate amount of a conventional adjuvant, for example, a Freund's complete adjuvant. After emulsification, the resulting emulsion is administered to each mammal several times every 4 to 21 days. Alternatively, an appropriate carrier may be used for the immunization with the sensitizing antigen.
  • After confirmation of a rise in the level of the desired antibody in the serum of the mammal thus immunized, immunocytes are collected from the mammal and subjected to cell fusion. Preferred examples of the immunocytes particularly include spleen cells.
  • Mammalian myeloma cells are used as partner parent cells to be fused with the immunocytes. Various cell lines known in the art, for example, P3U1 (P3-X63Ag8U1), P3 (P3x63Ag8.653) (J. Immunol. (1979) 123, 1548-1550), P3x63Ag8U.1 (Current Topics in Microbiology and Immunology (1978) 81, 1-7), NS-1 (Kohler. G. and Milstein. C. Eur. J. Immunol. (1976) 6, 511-519), MPC-1 (Margulies. D. H. et al., Cell (1976) 8, 405-415), SP2/0 (Shulman. M. et al., Nature (1978) 276, 269-270), FO (deSt. Groth, S. F. et al., J. Immunol. Methods (1980) 35, 1-21), S194 (Trowbridge, I. S. J. Exp. Med. (1978) 148, 313-323), R210 (Galfre, G. et al., Nature (1979) 277, 131-133), 240E-1, 240E-W and 240E-W2 are preferably used as the myeloma cells.
  • The cell fusion between the immunocytes and the myeloma cells can be carried out basically according to a method known in the art, for example, the method of Kohler and Milstein (Kohler, G. and Milstein, C. Methods Enzymol. (1981) 73, 3-46).
  • More specifically, the cell fusion is carried out, for example, in the presence of a cell fusion promoter in a conventional nutrient medium. For example, polyethylene glycol (PEG) or hemagglutinating virus of Japan (HVJ) is used as the fusion promoter. If desired, an auxiliary such as dimethyl sulfoxide may be further added for use in order to enhance fusion efficiency.
  • The ratio between the immunocytes and the myeloma cells used can be arbitrarily set. For example, an RPMII640 medium or a MEM medium suitable for the growth of the myeloma cell lines, or a conventional medium for use in this type of cell culture can be used as the medium for use in the cell fusion. In addition, a serum supplement such as fetal calf serum (FCS) may be used in combination with the medium.
  • For the cell fusion, the immunocytes and the myeloma cells are well mixed in a predetermined amount of the medium. A PEG solution (average molecular weight: for example, approximately 1000 to 6000) preheated to approximately 37° C. is usually added to the mixture at a concentration of 30 to 60% (w/v) and mixed therewith to form the hybridomas of interest. Subsequently, it is preferred to remove cell fusion agents or the like unfavorable for the growth of the hybridomas by repeating the procedures of sequentially adding an appropriate medium and removing the supernatant by centrifugation.
  • The hybridomas thus obtained are cultured in a conventional selective medium, for example, a HAT medium (medium containing hypoxanthine, aminopterin, and thymidine) for selection. This culture in the HAT medium is continued for a period (usually, several days to several weeks) sufficient for the death of cells (non-fused cells) other than the hybridomas of interest. Subsequently, hybridomas producing the antibody of interest are screened for and cloned as single clones by a conventional limiting dilution method.
  • In addition to such obtainment of the hybridomas by the immunization of non-human animals with the antigen, hybridomas producing human antibodies having the desired activity (e.g., cell growth inhibitory activity) may be obtained by sensitizing human lymphocytes, for example, EB virus-infected human lymphocytes, with the protein, protein-expressing cells, or lysates thereof in vitro and fusing the sensitized lymphocytes with human-derived myeloma cells capable of dividing permanently, for example, U266 (Registration No. TIB196).
  • The monoclonal antibody-producing hybridomas thus prepared can be subcultured in a conventional medium and can also be stored for a long period in liquid nitrogen.
  • Specifically, the desired antigen or cells expressing the desired antigen are used as a sensitizing antigen in immunization according to a conventional immunization method. The obtained immunocytes are fused with parent cells known in the art according to a conventional cell fusion method. Monoclonal antibody-producing cells (hybridomas) can be screened for by a conventional screening method to prepare the antibody of interest.
  • The antigen can be prepared according to, for example, a method using animal cells (JP Patent Publication (Kohyo) No. 2007-530068 A (2007)) or a method using baculovirus (e.g., International Publication No. WO98/46777). When the antigen has low immunogenicity, this antigen can be bound to an immunogenic macromolecule such as albumin for immunization. The antigen may be administered together with an adjuvant for immunization.
  • Alternatively, the antibody of the present invention may be obtained as a gene recombinant antibody produced using a gene recombination technique which involves: cloning a gene of the antibody from a hybridoma: incorporating the antibody gene into appropriate vectors, and transferring the vectors into hosts (see, e.g., Carl, A. K. Borrebaeck, James, W. Larrick, THERAPEUTIC MONOCLONAL ANTIBODIES, Published in the United Kingdom by MACMILLAN PUBLISHERS LTD, 1990). Specifically, variable region (V region) cDNAs of the antibody are synthesized from the mRNAs of the hybridoma using reverse transcriptase. After obtainment of DNAs encoding the V regions of the antibody of interest, the DNAs are ligated with DNAs encoding the desired antibody constant regions (C regions). The resulting ligation products are incorporated into expression vectors. Alternatively, the antibody V region-encoding DNAs may be incorporated into expression vectors containing antibody C region DNAs. These DNAs are incorporated into the expression vectors so as to be expressed under the control of expression control regions, for example, an enhancer and a promoter. Next, host cells can be transformed with the resulting expression vectors and allowed to express the antibody.
  • The anti-CAPRIN-1 antibody of the present invention is preferably a monoclonal antibody. The monoclonal antibody includes human monoclonal antibodies, non-human animal monoclonal antibodies (e.g., mouse, rat, rabbit, and chicken monoclonal antibodies), chimeric monoclonal antibodies, and the like. The monoclonal antibody may be prepared by culturing hybridomas obtained by the fusion between spleen cells from non-human mammals (e.g., mice, human antibody-producing mice, chickens, or rabbits) immunized with the CAPRIN-1 protein or a fragment thereof and myeloma cells. The chimeric antibody is an antibody prepared from a combination of sequences derived from different animals and is, for example, an antibody composed of heavy chain and light chain variable regions of a mouse antibody and heavy chain and light chain constant regions of a human antibody. The chimeric antibody can be prepared using a method known in the art and is obtained, for example by: ligating DNAs encoding the antibody V regions with DNAs encoding the human antibody C regions; incorporating the resulting ligation products into expression vectors; and transferring the vectors into hosts for antibody production.
  • In Examples mentioned later, a plurality of humanized monoclonal antibodies and a human-rabbit chimeric monoclonal antibody were prepared and confirmed to have a strong antitumor effect. All of these monoclonal antibodies have a heavy chain variable region (VH region) comprising CDR1 represented by the amino acid sequence of SEQ ID NO: 1, CDR2 represented by the amino acid sequence of SEQ ID NO: 2, and CDR3 represented by the amino acid sequence of SEQ ID NO: 3, and a light chain variable region (VL region) comprising CDR1 represented by the amino acid sequence of SEQ ID NO: 4, CDR2 represented by the amino acid sequence of SEQ ID NO: 5, and CDR3 represented by the amino acid sequence of SEQ ID NO: 6. These monoclonal antibodies include humanized antibody #0 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 11, humanized antibody #1 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 11, humanized antibody #2 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 12, humanized antibody #3 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 13, humanized antibody #4 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 12, humanized antibody #5 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 13, humanized antibody #6 consisting of a VH region having the amino acid sequence of SEQ ID NO: 7 and a VL region having the amino acid sequence of SEQ ID NO: 15, humanized antibody #7 consisting of a VH region having the amino acid sequence of SEQ ID NO: 8 and a VL region having the amino acid sequence of SEQ ID NO: 15, humanized antibody #8 consisting of a VH region having the amino acid sequence of SEQ ID NO: 9 and a VL region having the amino acid sequence of SEQ ID NO: 15, humanized antibody #9 consisting of a VH region having the amino acid sequence of SEQ ID NO: 10 and a VL region having the amino acid sequence of SEQ ID NO: 14, humanized antibody #10 consisting of a VH region having the amino acid sequence of SEQ ID NO: 10 and a VL region having the amino acid sequence of SEQ ID NO: 15, and a human-rabbit chimeric antibody consisting of a VH region having the amino acid sequence of SEQ ID NO: 20 and a VL region having the amino acid sequence of SEQ ID NO: 21.
  • The humanized antibody, also called reshaped human antibody, is an engineered antibody. The humanized antibody is constructed by grafting complementarity-determining regions of a human antibody with complementarity-determining regions of an antibody derived from an immunized animal. A general gene recombination approach therefor is also known.
  • Specifically, DNA sequences designed so as to link complementarity-determining regions of, for example, a mouse, rabbit, or chicken antibody, and framework regions of a human antibody are synthesized by PCR from several prepared oligonucleotides having terminal portions overlapping with each other. The obtained DNAs are ligated with DNAs encoding human antibody constant regions. Subsequently, the resulting ligation products are incorporated into expression vectors, which are then transferred to hosts for antibody production to obtain the antibody of interest (see European Patent Application Publication No. EP239400 and International Publication No. WO96/02576). The framework regions of a human antibody connected via the complementarity-determining regions are selected such that the complementarity-determining regions form a favorable antigen-binding site. If necessary, amino acids in the framework regions of antibody variable regions may be substituted such that the complementarity-determining regions of the resulting reshaped human antibody form an appropriate antigen-binding site (Sato K. et al., Cancer Research 1993, 53: 851-856). In addition, these framework regions may be replaced with framework regions derived from various human antibodies (see International Publication No. WO99/51743).
  • For preparing the chimeric antibody or the humanized antibody, amino acids in variable regions (e.g., FRs) or constant regions may be substituted, for example, by other amino acids.
  • The amino acid substitution is the substitution of one or more, for example, less than 15, less than 10, 8 or less, 7 or less, 6 or less, 5 or less, 4 or less, 3 or less, or 2 or less amino acids, preferably 1 to 9 amino acids. The substituted antibody should be functionally equivalent to an unsubstituted antibody.
  • In this context, the phrase “functionally equivalent” means that an antibody concerned has biological or biochemical activity similar to that of the antibody of the present invention, specifically, the antibody concerned has the function of damaging tumor and essentially causes no rejection when applied to humans, for example. Examples of such activity can include cell growth inhibitory activity and binding activity.
  • An amino acid substitution method which involves introducing a mutation into a polypeptide is well known to those skilled in the art as a method for preparing a polypeptide functionally equivalent to a certain polypeptide. For example, those skilled in the art can appropriately introduce amino acid substitution into the antibody of the present invention using site-directed mutagenesis (Hashimoto-Gotoh, T. et al., (1995) Gene 152, 271-275; Zoller, M J., and Smith, M. (1983) Methods Enzymol. 100, 468-500; Kramer, W. et al., (1984) Nucleic Acids Res. 12, 9441-9456: Kramer, W. and Fritz, H J., (1987) Methods Enzymol. 154, 350-367; Kunkel. TA., (1985) Proc. Natl. Acad. Sci. USA. 82, 488-492: Kunkel (1988) Methods Enzymol. 85, 2763-2766) or the like, thereby preparing an antibody functionally equivalent to the antibody of the present invention.
  • In the case of introducing amino acid substitution, the substitution is desirably conservative amino acid substitution. The conservative amino acid substitution is the substitution between amino acids similar in properties such as charge, side chains, polarity, and aromaticity. The amino acids can be classified in terms of similar properties into, for example: basic amino acids (arginine, lysine, and histidine); acidic amino acids (aspartic acid and glutamic acid); uncharged polar amino acids (glycine, asparagine, glutamine, serine, threonine, cysteine, and tyrosine); nonpolar amino acids (leucine, isoleucine, alanine, valine, proline, phenylalanine, tryptophan, and methionine): branched amino acids (leucine, valine, and isoleucine); and aromatic amino acids (phenylalanine, tyrosine, tryptophan, and histidine).
  • Examples of modified antibodies can include antibodies bound with various molecules such as polyethylene glycol (PEG). In the modified antibody of the present invention, the substance to be bound is not limited. In order to obtain such a modified antibody, the obtained antibody can be chemically modified. A method therefor has already been established in the art.
  • The antibody that recognizes a CAPRIN-1 protein or a CAPRIN-1 fragment polypeptide can be obtained by a method generally known to those skilled in the art. The antibody can be obtained by, for example, a method which involves determining an epitope of the CAPRIN-1 protein recognized by an anti-CAPRIN-1 antibody by a conventional method (e.g., epitope mapping or an epitope identification method mentioned later) and preparing an antibody using a polypeptide having an amino acid sequence contained in the epitope as an immunogen, or a method which involves determining an epitope for an antibody prepared by a conventional method and selecting an antibody that recognizes the same epitope as that for an anti-CAPRIN-1 antibody.
  • The antibody of the present invention is an antibody having immunological reactivity with CAPRIN-1, an antibody that specifically recognizes CAPRIN-1, or an antibody that specifically binds to CAPRIN-1 and exhibits cytotoxic activity against a cancer or a tumor growth inhibitory effect. It is preferred that the antibody should be an antibody having a structure so as to cause little or no rejection in recipient animals. Examples of such antibodies include human antibodies, humanized antibodies, chimeric antibodies (e.g., human-rabbit chimeric antibodies), single-chain antibodies, and bispecific antibodies when the recipient animals are humans. These antibodies are recombinant antibodies having heavy chain and light chain variable regions derived from a human antibody, having heavy chain and light chain variable regions comprising complementarity-determining regions (CDR1, CDR2, and CDR3) derived from a non-human animal antibody and framework regions (FR1, FR2, FR3, and FR4) derived from a human antibody, or having heavy chain and light chain variable regions derived from a non-human animal antibody and heavy chain and light chain constant regions derived from a human antibody. Preferred antibodies are the former two antibodies.
  • These recombinant antibodies can be prepared as follows: a DNA encoding the monoclonal antibody (e.g., a human, mouse, rat, rabbit, or chicken monoclonal antibody) against human CAPRIN-1 is cloned from antibody-producing cells such as hybridomas, and this is used as a template in RT-PCR or the like to prepare DNAs encoding the light chain and heavy chain variable regions of the antibody. The respective sequences of the light chain and heavy chain variable regions, the respective sequences of CDR1, CDR2, and CDR3 in each region, or the respective sequences of FR1, FR2, FR3, and FR4 in each region can be determined on the basis of, for example, the Kabat EU numbering system (Kabat et al., Sequences of Proteins of Immunological Interest, 5th Ed. Public Health Service, National Institute of Health, Bethesda, Md. (1991)).
  • Such a DNA encoding each of these variable regions or a DNA encoding each complementarity-determining region is further prepared using a gene recombination technique (Sambrook et al., Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)) or a DNA synthesizer. In this context, the human monoclonal antibody-producing hybridomas can be prepared by immunizing human antibody-producing animals (e.g., mice) with human CAPRIN-1 and then fusing spleen cells excised from the immunized animals with myeloma cells. Aside from this, DNAs encoding human antibody-derived light chain or heavy chain variable and constant regions are prepared, if necessary, using a gene recombination technique or a DNA synthesizer.
  • For the humanized antibody, the CDR coding sequences in the DNA encoding the human antibody-derived light chain or heavy chain variable region can be substituted by corresponding CDR coding sequences of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody to thereby prepare a humanized antibody-encoding DNA. In the case of, for example, a humanized antibody in which CDR coding sequences derived from a human antibody is substituted by corresponding CDR coding sequences derived from a mouse antibody, each variable region is constituted by human FR1, mouse CDR1, human FR2, mouse CDR2, human FR3, mouse CDR4, and human FR4 in this order from the N-terminus.
  • For the chimeric antibody, the DNA encoding the light chain or heavy chain variable region of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody can be ligated with the DNA encoding a light chain or heavy chain constant region derived from a human antibody to prepare a chimeric antibody-encoding DNA.
  • In the case of the single-chain antibody, this antibody refers to an antibody comprising a heavy chain variable region and a light chain variable region linearly linked via a linker. A DNA encoding the single-chain antibody can be prepared by ligating a DNA encoding the heavy chain variable region, a DNA encoding the linker, and a DNA encoding the light chain variable region. In this context, both of the heavy chain variable region and the light chain variable region are derived from a human antibody or derived from a human antibody having complementarity-determining regions alone substituted by complementarity-determining regions of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody. The linker consists of 12 to 19 amino acids. Examples thereof include (G4S)3 consisting of 15 amino acids (G.-B. Kim et al., Protein Engineering Design and Selection 2007, 20 (9): 425-432).
  • In the case of the bispecific antibody (e.g., diabody), this antibody refers to an antibody capable of specifically binding to two different epitopes. A DNA encoding the bispecific antibody can be prepared by ligating, for example, a DNA encoding heavy chain variable region A, a DNA encoding light chain variable region B, a DNA encoding heavy chain variable region B, and a DNA encoding light chain variable region A in this order (provided that the DNA encoding a light chain variable region B and the DNA encoding a heavy chain variable region B are ligated via a DNA encoding a linker as described above). In this context, all of the heavy chain variable regions and the light chain variable regions are derived from a human antibody or derived from a human antibody having complementarity-determining regions alone substituted by complementarity-determining regions of a non-human animal (e.g., mouse, rat, rabbit, or chicken)-derived antibody.
  • The recombinant DNAs thus prepared can be incorporated into one or more appropriate vectors, which are then transferred to host cells (e.g., mammalian cells, yeast cells, and insect cells) so that the DNAs are (co)expressed to produce the recombinant antibody of interest (P. J. Delves., ANTIBODY PRODUCTION ESSENTIAL TECHNIQUES., 1997 WILEY: P. Shepherd and C. Dean., Monoclonal Antibodies., 2000 OXFORD UNIVERSITY PRESS; J. W. Goding., Monoclonal Antibodies: principles and practice., 1993 ACADEMIC PRESS).
  • Examples of the antibody of the present invention prepared by any of the methods described above include the following antibodies (a) to (1) comprising a heavy chain variable region comprising SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising SEQ ID NOs: 4, 5, and 6, obtained in Examples mentioned later:
  • (a) an antibody constituted by a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 15,
  • (b) an antibody constituted by a heavy chain variable region of SEQ ID NO: 10 and a light chain variable region of SEQ ID NO: 15.
  • (c) an antibody constituted by a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 15,
  • (d) an antibody constituted by a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 13,
  • (e) an antibody constituted by a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 12,
  • (f) an antibody constituted by a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12,
  • (g) an antibody constituted by a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 13,
  • (h) an antibody constituted by a heavy chain variable region of SEQ ID NO: 10 and a light chain variable region of SEQ ID NO: 14,
  • (i) an antibody constituted by a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 11,
  • (j) an antibody constituted by a heavy chain variable region of SEQ ID NO: 7 and a light chain variable region of SEQ ID NO: 11,
  • (k) an antibody constituted by a heavy chain variable region of SEQ ID NO: 9 and a light chain variable region of SEQ ID NO: 15, and
  • (l) an antibody constituted by a heavy chain variable region of SEQ ID NO: 20 and a light chain variable region of SEQ ID NO: 21.
  • In this context, the amino acid sequences represented by SEQ ID NOs: 1, 2, and 3 correspond to CDR1, CDR2, and CDR3, respectively, of the heavy chain variable region of the rabbit antibody, and the amino acid sequences represented by SEQ ID NOs: 4, 5, and 6 correspond to CDR1, CDR2, and CDR3, respectively, of the light chain variable region of the rabbit antibody.
  • The humanized antibody, the chimeric antibody, the single-chain antibody, or the bispecific antibody of the present invention is, for example, any of the following antibodies (i) to (xiv):
  • (i) an antibody comprising a heavy chain variable region comprising the amino acid sequences of SEQ ID NOs: 1, 2, and 3 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof, and a light chain variable region comprising the amino acid sequences of SEQ ID NOs: 4, 5, and 6 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof,
  • (ii) an antibody comprising a heavy chain variable region comprising the amino acid sequences of SEQ ID NOs: 1, 2, and 3 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequences of SEQ ID NOs: 4, 5, and 6 and the amino acid sequences of human antibody-derived framework regions, or substituted forms thereof and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (iii) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (iv) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence.
  • (v) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (vi) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 13 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (vii) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (viii) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 12 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (ix) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 13 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (x) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 10 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 14 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (xi) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 8 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 11 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (xii) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 7 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 11 and a light chain constant region comprising a human antibody-derived amino acid sequence,
  • (xiii) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 9 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 15 and a light chain constant region comprising a human antibody-derived amino acid sequence, and
  • (xiv) an antibody comprising a heavy chain variable region comprising the amino acid sequence of SEQ ID NO: 20 and a heavy chain constant region comprising a human antibody-derived amino acid sequence, and a light chain variable region comprising the amino acid sequence of SEQ ID NO: 21 and a light chain constant region comprising a human antibody-derived amino acid sequence.
  • The sequences of framework regions of the human antibody heavy chain and light chain constant and variable regions are available from, for example, NCBI (USA; GenBank, UniGene, etc.). For example, the following sequences can be referred to: Registration No. J00228 for a human IgG1 heavy chain constant region; Registration No. J00230 for a human IgG2 heavy chain constant region; Registration No. X03604 for a human IgG3 heavy chain constant region: Registration No. K01316 for a human IgG4 heavy chain constant region; Registration Nos. V00557, X64135, X64133, etc., for a human light chain κ constant region: and Registration Nos. X64132. X64134, etc., for a human light chain λ constant region.
  • Preferably, these antibodies have cytotoxic activity and can thereby exert an antitumor effect (or antitumor activity).
  • The aforementioned antibodies may each have the substitution, deletion, or addition of one or several amino acids in a complementarity-determining region sequence, a framework region sequence, and/or a constant region sequence, as long as the resulting antibody has such specificity that it can specifically recognize CAPRIN-1. In this context, the term “several” means preferably 1 to 9.
  • The affinity constant Ka (kon/koff) of the antibody of the present invention for a CAPRIN-1 protein or a fragment thereof is preferably at least 5×108 M−1, at least 109 M−1, at least 5×109 M−1, at least 1010 M−1, at least 5×1010 M−1, at least 1011 M−1, at least 5×1011 M−1, at least 1012 M−1, at least 1013 M−1, or at least 1014 M−1,
  • One mechanism underlying the antitumor effect of the antibody of the present invention on CAPRIN-1-expressing cancer cells is the antibody-dependent cellular cytotoxicity (ADCC) of effector cells against the CAPRIN-1-expressing cells. The antitumor activity of the antibody of the present invention through ADCC can be enhanced by substituting one or several amino acids in the heavy chain constant region of the antibody of the present invention or by removing fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain attached to the heavy chain constant region. The antitumor activity of the antibody of the present invention through ADCC can be further enhanced by combining the amino acid substitution and fucose removal of the heavy chain constant region.
  • Such an antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain attached to the heavy chain constant region according to the present invention may be used alone or may be a composition with a fucosylated antibody. It is preferred that the antibody composition should be composed mainly of the antibody lacking fucose.
  • The antibody in which one or several amino acids in the heavy chain constant region are substituted can be prepared with reference to, for example, International Publication No. WO2004/063351, International Publication No. WO2011/120135, U.S. Pat. No. 8,388,955, International Publication No. WO2011/005481, U.S. Pat. No. 6,737,056, and International Publication No. WO2005/063351. The antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain in the heavy chain constant region, or cells producing the antibody can be prepared with reference to, for example, U.S. Pat. No. 6,602,684, European Patent No. 1914244, and U.S. Pat. No. 7,579,170. The composition of the antibody lacking fucose added to N-acetylglucosamine in a N-glycoside-linked sugar chain attached to the heavy chain constant region, and the fucosylated antibody, or cells producing the composition can be prepared with reference to, for example, U.S. Pat. No. 8,642,292.
  • The antibody of the present invention can be conjugated with an antitumor agent. The conjugation of the antibody with the antitumor agent can be carried out via a spacer having a group (e.g., a succinimidyl group, a formyl group, a 2-pyridyldithio group, a maleimidyl group, an alkoxycarbonyl group, or a hydroxy group) reactive with an amino group, a carboxyl group, a hydroxy group, a thiol group, or the like.
  • Examples of the antitumor agent include the following antitumor agents publicly known in literatures, etc.: paclitaxel, doxorubicin, daunorubicin, cyclophosphamide, methotrexate, 5-fluorouracil, thiotepa, busulfan, improsulfan, piposulfan, benzodopa, carboquone, meturedopa, uredopa, altretamine, triethylenemelamine, triethylenephosphoramide, triethylenethiophosphoramide, trimethylolomelamine, bullatacin, bullatacinone, camptothecin, bryostatin, callystatin, cryptophycin 1, cryptophycin 8, dolastatin, duocarmycin, eleutherobin, pancratistatin, sarcodictyin, spongistatin, chlorambucil, chlomaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard, carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine, calicheamicin, dynemicin, clodronate, esperamicin, aclacinomycin, actinomycin, authramycin, azaserine, bleomycin, cactinomycin, carabicin, carminomycin, carzinophilin, chromomycin, dactinomycin, detorbicin, 6-diazo-5-oxo-L-norleucine, Adriamycin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycin C, mycophenolic acid, nogalamycin, olivomycin, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin, denopterin, pteropterin, trimetrexate, fludarabine, 6-mercaptopurine, thiamiprine, thioguanine, ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens (e.g., calusterone, dromostanolone propionate, epitiostanol, mepitiostane, and testolactone), aminoglutethimide, mitotane, trilostane, frolinic acid, aceglatone, aldophosphamide glycoside, aminolevulinic acid, eniluracil, amsacrine, bestrabucil, bisantrene, edatraxate, defofamine, demecolcine, diaziquone, elfomithine, elliptinium acetate, epothilone, etoglucid, lentinan, lonidamine, maytansine, ansamitocin, mitoguazone, mitoxantrone, mopidanmol, nitraerine, pentostatin, phenamet, pirarubicin, losoxantrone, podophyllinic acid, 2-ethylhydrazide, procarbazine, razoxane, rhizoxin, schizophyllan, spirogermanium, tenuazonic acid, triaziquone, roridin A, anguidine, urethane, vindesine, dacarbazine, mannomustine, mitobronitol, mitolactol, pipobroman, gacytosine, docetaxel, chlorambucil, gemcitabine, 6-thioguanine, mercaptopurine, cisplatin, oxaliplatin, carboplatin, vinblastine, etoposide, ifosfamide, mitoxantrone, vincristine, vinorelbine, novantrone, teniposide, edatrexate, daunomycin, aminopterin, Xeloda, ibandronate, irinotecan, topoisomerase inhibitors, difluoromethylomithine (DMFO), retinoic acid, capecitabine, and pharmaceutically acceptable salts and derivatives thereof
  • When the antibody is an antibody conjugated with an antitumor agent, a method for evaluating whether to exert antitumor activity can involve, for example, for the mouse-derived anti-CAPRIN-1 antibody, reacting a drug-attached secondary antibody binding to a mouse antibody together therewith to evaluate ex vivo the antitumor effect on human cancer cells. This evaluation can be conducted using, for example, an anti-human IgG antibody (Hum-ZAP (Advanced Targeting Systems, Inc.)), which is a second immunotoxin bound with saporin.
  • Alternatively, the antibody of the present invention can be administered in combination with an antitumor agent to thereby produce a higher therapeutic effect. This approach is adaptable to a patient with a CAPRIN-1-expressing cancer either before or after surgical treatment. This approach can be applied, particularly after surgery, to a CAPRIN-1-expressing cancer, which has been treated conventionally with an antitumor agent alone, to produce higher prevention of cancer recurrence or prolongation of survival time.
  • For example, any of the antitumor agents described above can be used as the antitumor agent for use in the combined administration with the antibody of the present invention. Particularly, cyclophosphamide, paclitaxel, docetaxel, or vinorelbine is preferably used.
  • Alternatively, the antibody of the present invention may be bound to a radioisotope publicly known in literatures, etc., such as 211At, 131I, 125I, 90Y, 186Re, 188Re, 153SM, 212Bi, 32P, 175Lu, 176Lu, 89Sr, 64Cu, or 111In (Hideo Saji, YAKUGAKU ZASSHI 128 (3) 323-332, 8 (2008), Jpn). A radioisotope effective for the treatment or diagnosis of tumor is desirable. Such a radioisotope is also included in the antitumor agent according to the present invention.
  • <Antitumor Effect>
  • The antitumor effect of the anti-CAPRIN-1 antibody used in the present invention on CAPRIN-1-expressing cancer cells is considered to take place under the following mechanism or the like: the antibody-dependent cellular cytotoxicity (ADCC) of effector cells against the CAPRIN-1-expressing cells mentioned above and the antibody-dependent cellular phagocytosis (ADCP) of the CAPRIN-1-expressing cells. However, the scope of the present invention is not intended to be limited by this mechanism.
  • Thus, the activity of the anti-CAPRIN-1 antibody used in the present invention can be evaluated, as specifically shown below in Examples, by measuring ex vivo the ADCC activity or the ADCP activity against CAPRIN-1-expressing cancer cells.
  • The anti-CAPRIN-1 antibody used in the present invention binds to the CAPRIN-1 protein on cancer cells and exhibits an antitumor effect through the activity or the like. Thus, the anti-CAPRIN-1 antibody of the present invention is presumably useful in the treatment or prevention of a cancer. Specifically, the present invention provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising the anti-CAPRIN-1 antibody as an active ingredient. The anti-CAPRIN-1 antibody used for the purpose of administration to human bodies (antibody therapy) is preferably a human antibody or a humanized antibody for reducing immunogenicity.
  • An anti-CAPRIN-1 antibody with higher binding affinity for the CAPRIN-1 protein on the surface of cancer cells exerts stronger antitumor activity. Thus, the antibody of the present invention has high binding affinity for the CAPRIN-1 protein and can therefore be expected to have a stronger antitumor effect. Accordingly, the antibody of the present invention is adaptable to a pharmaceutical composition intended for the treatment and/or prevention of a cancer. Such high binding affinity of the antibody of the present invention is preferably at least 5×108 M−1, at least 109 M−1, at least 5×109 M−1, at least 1010 M−1, at least 5×1010 M−1, at least 1011 M−1, at least 5×1011 M−1, at least 1012 M−1, at least 1013 M−1, or at least 1014 M|1, in terms of an association constant (affinity constant) Ka (kon/koff), as described above.
  • <Binding to Antigen-Expressing Cell>
  • The ability of the antibody to bind to CAPRIN-1 can be determined by use of binding assay using, for example, ELISA, Western blot, immunofluorescence, and flow cytometry analysis, as described in Examples.
  • <Immunohistochemical Staining>
  • The antibody that recognizes CAPRIN-1 can be used in immunohistochemistry by a method well known to those skilled in the art. The antibody that recognizes CAPRIN-1 can be tested for its reactivity with CAPRIN-1, for example, using a paraformaldehyde- or acetone-fixed frozen section or a paraformaldehyde-fixed and paraffin-embedded section of a tissue obtained from a patient during surgical treatment or a tissue obtained from an animal carrying a xenograft tissue inoculated with a cell line expressing CAPRIN-1 either spontaneously or after transfection.
  • For immunohistochemical staining, the antibody reactive with CAPRIN-1 can be stained by various methods. For example, the antibody can be visualized through reaction with a horseradish peroxidase-conjugated goat anti-mouse antibody, goat anti-rabbit antibody, or goat anti-chicken antibody.
  • <Pharmaceutical Composition and Method for Treating and/or Preventing Cancer>
  • The target of the pharmaceutical composition for the treatment and/or prevention of a cancer of the present invention is not particularly limited as long as the target is cancer (cells) expressing a CAPRIN-1 gene.
  • The terms “tumor” and “cancer” used in the present specification mean malignant neoplasm and are used interchangeably with each other.
  • The cancer targeted in the present invention may be any cancer expressing the CAPRIN-1 protein on the cell membrane surface. The cancer is preferably breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer as mentioned above.
  • More specifically, examples of these cancers include, but are not limited to, breast adenocarcinoma, complex-type breast adenocarcinoma, malignant mixed tumor of the mammary gland, intraductal papillary adenocarcinoma, lung adenocarcinoma, squamous cell cancer, small-cell cancer, large-cell cancer, glioma which is tumor of neuroepithelial tissue, glioblastoma, neuroblastoma, ependymoma, neuronal tumor, embryonal neuroectodermal tumor, neurilemmoma, neurofibroma, meningioma, chronic lymphocytic leukemia, gastrointestinal lymphoma, alimentary lymphoma, small to medium cell-type lymphoma, cecal cancer, ascending colon cancer, descending colon cancer, transverse colon cancer, sigmoid colon cancer, rectal cancer, epithelial ovarian cancer, germ cell tumor, stromal cell tumor, pancreatic ductal carcinoma, invasive pancreatic ductal carcinoma, pancreatic adenocarcinoma, acinar cell carcinoma, adenosquamous carcinoma, giant cell tumor, intraductal papillary-mucinous neoplasm, mucinous cystic neoplasm, pancreatoblastoma, islet-cell adenoma. Frants tumor, serous cystadenocarcinoma, solid-pseudopapillary tumor, gastrinoma, glucagonoma, insulinoma, multiple endocrine neoplasia type-1 (Wermer's syndrome), nonfunctional islet cell tumor, somatostatinoma, VIPoma, uterine cervix cancer, uterine body cancer, fibrosarcoma, sarcoma of bones or joints, Ewing's sarcoma. Wilms tumor, hepatoblastoma, soft tissue sarcoma, acute leukemia, chronic leukemia, spinal cord tumor, malignant soft tissue tumor, teratoma group tumor, and head and neck cancer including hypopharynx cancer, oropharynx cancer, tongue cancer, epipharynx cancer, oral cancer, lip cancer, sinus cancer, and throat cancer.
  • The recipient subjects (patients) are preferably mammals, for example, mammals including primates, pet animals, livestock, and sport animals and are particularly preferably humans, dogs, and cats.
  • In the case of using the antibody of the present invention as a pharmaceutical composition, the pharmaceutical composition can be formulated by a method generally known to those skilled in the art. For example, the pharmaceutical composition can be used in the form of a parenteral injection of an aseptic solution or suspension with water or any other pharmaceutically acceptable liquid. For example, the pharmaceutical composition may be formulated with the antibody mixed in a unit dosage form required for generally accepted pharmaceutical practice, in appropriate combination with pharmacologically acceptable carriers or media, specifically, sterilized water, physiological saline, a plant oil, an emulsifier, a suspending agent, a surfactant, a stabilizer, a flavoring agent, an excipient, a vehicle, a preservative, a binder, etc. The amount of the active ingredient in such a preparation is determined such that an appropriate dose within the prescribed range can be achieved.
  • An aseptic composition for injection can be formulated according to conventional pharmaceutical practice using a vehicle such as injectable distilled water.
  • Examples of aqueous solutions for injection include physiological saline, isotonic solutions containing glucose and other adjuvants, for example, D-sorbitol, D-mannose, D-mannitol, and sodium chloride. These solutions may be used in combination with an appropriate solubilizer, for example, an alcohol (specifically, ethanol) or a polyalcohol (e.g., propylene glycol and polyethylene glycol), or a nonionic surfactant, for example, Polysorbate 80™ or HCO-60.
  • Examples of oily solutions include sesame oil and soybean oil. These solutions may be used in combination with benzyl benzoate or benzyl alcohol as a solubilizer. The solutions may be further mixed with a buffer (e.g., a phosphate buffer solution and a sodium acetate buffer solution), a soothing agent (e.g., procaine hydrochloride), a stabilizer (e.g., benzyl alcohol and phenol), and an antioxidant. The injection solutions thus prepared are usually charged into appropriate ampules.
  • The pharmaceutical composition of the present invention is administered orally or parenterally, preferably parenterally. Specific examples of its dosage forms include injections, intranasal administration agents, transpulmonary administration agents, and percutaneous administration agents. Examples of the injections include intravenous injection, intramuscular injection, intraperitoneal injection, and subcutaneous injection, through which the pharmaceutical composition can be administered systemically or locally.
  • Also, the administration method can be appropriately selected depending on the age, weight, sex, symptoms, etc., of a patient. The dose of a pharmaceutical composition containing the antibody or a polynucleotide encoding the antibody can be selected within a range of, for example, 0.0001 to 10(X) mg/kg of body weight per dose. Alternatively, the dose can be selected within a range of, for example, 0.001 to (X00000 mg/body of a patient, though the dose is not necessarily limited to these numeric values. Although the dose and the administration method vary depending on the weight, age, sex, symptoms, etc., of a patient, those skilled in the art can appropriately select the dose and the method.
  • The pharmaceutical composition comprising the antibody of the present invention or the fragment thereof can be administered to a subject to treat and/or prevent a cancer, preferably breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor. Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
  • The present invention further encompasses a method for treating and/or preventing a cancer, comprising administering the pharmaceutical composition of the present invention in combination with the antitumor agent as exemplified above or a pharmaceutical composition comprising the antitumor agent to a subject. The antibody of the present invention or the fragment thereof may be administered simultaneously with or separately from the antitumor agent to the subject. In the case of separate administration, either of their pharmaceutical compositions may be administered first or later. Their dosing intervals, doses, administration routes, and the number of doses can be appropriately selected by a specialist. In the case of simultaneous administration, the dosage form also includes, for example, a pharmaceutical composition formulated by mixing the antibody of the present invention or the fragment thereof and the antitumor agent in a pharmacologically acceptable carrier (or medium). The above descriptions about prescription, formulation, administration routes, doses, cancers, etc., as to the pharmaceutical compositions and dosage forms containing the antibody of the present invention are also applicable to any of the pharmaceutical compositions and dosage forms containing the antitumor agent.
  • Thus, the present invention also provides a combination drug product for the treatment and/or prevention of a cancer, comprising the pharmaceutical composition of the present invention and a pharmaceutical composition comprising the antitumor agent as exemplified above, and a method for treating and/or preventing a cancer, comprising administering the combination drug product. The present invention also provides a pharmaceutical composition for the treatment and/or prevention of a cancer, comprising the antibody of the present invention or the fragment thereof and the antitumor agent together with a pharmacologically acceptable carrier.
  • <Polypeptide and DNA>
  • The present invention further provides a DNA encoding the antibody of the present invention, a DNA encoding the heavy chain or the light chain of the antibody, and a DNA encoding the heavy chain or light chain variable region of the antibody. Such DNAs include, in the case of the antibody (a), for example, a DNA encoding a heavy chain variable region, comprising nucleotide sequences encoding the amino acid sequences of SEQ ID NOs: 1, 2, and 3, and a DNA encoding a light chain variable region, comprising nucleotide sequences encoding the amino acid sequences of SEQ ID NOs: 4, 5, and 6.
  • Since the complementarity-determining regions encoded by the DNAs having these sequences are regions that determine the specificity of the antibody, sequences encoding the other regions (i.e., constant regions and framework regions) of the antibody may be sequences derived from a different antibody. In this context, the different antibody also includes an antibody derived from a non-human organism, but is preferably one derived from a human from the viewpoint of reduction in adverse reaction. Specifically, for the DNAs mentioned above, it is preferred that regions encoding the respective framework regions of the heavy chain and the light chain and each constant region should comprise nucleotide sequences encoding the corresponding amino acid sequences derived from a human antibody.
  • Further examples of the DNA encoding the antibody of the present invention include a DNA encoding a heavy chain variable region, comprising a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 8, and a DNA in which a region encoding a light chain variable region comprises a nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 15. In this context, an example of the nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 8 is the nucleotide sequence of SEQ ID NO: 23. Also, an example of the nucleotide sequence encoding the amino acid sequence of SEQ ID NO: 15 is the nucleotide sequence of SEQ ID NO: 30. For these DNAs, it is also preferred that regions encoding the respective constant regions of the heavy chain and the light chain should comprise nucleotide sequences encoding the corresponding amino acid sequences derived from a human antibody.
  • These antibody DNAs can be obtained by, for example, the method mentioned above or the following method: first, total RNA is prepared from the hybridoma related to the antibody of the present invention using a commercially available RNA extraction kit, and cDNA is synthesized with reverse transcriptase using random primers or the like. Subsequently, the cDNA encoding the antibody is amplified by PCR using, as primers, oligonucleotides having sequences respectively conserved in the respective variable regions of a known mouse antibody heavy chain gene and light chain gene. Sequences encoding constant regions can be obtained by amplifying known sequences by PCR. The nucleotide sequence of the resulting DNA can be determined by a routine method, for example, by integration into a plasmid or a phage for sequencing.
  • The present invention further provides polypeptides and DNAs described in the following (i) to (xv) related to the antibodies (i) to (xiv):
  • (i) a heavy chain CDR polypeptide selected from the group consisting of the amino acid sequences represented by SEQ ID NOs: 1, 2, and 3, and a DNA encoding the polypeptide,
  • (ii) a light chain CDR polypeptide selected from the amino acid sequences represented by SEQ ID NOs: 4, 5, and 6, and a DNA encoding the polypeptide,
  • (iii) a polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 30, respectively,
  • (iv) a polypeptide comprising the amino acid sequences of SEQ ID NO: 10 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 25 or SEQ ID NO: 30, respectively,
  • (v) a polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 30, respectively,
  • (vi) a polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 13, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 28, respectively.
  • (vii) a polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 12, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 27, respectively,
  • (viii) a polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 12, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 27, respectively,
  • (ix) a polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 13, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 28, respectively,
  • (x) a polypeptide comprising the amino acid sequences of SEQ ID NO: 10 or SEQ ID NO: 14, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 25 or SEQ ID NO: 29, respectively,
  • (xi) a polypeptide comprising the amino acid sequences of SEQ ID NO: 8 or SEQ ID NO: 11, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 23 or SEQ ID NO: 26, respectively.
  • (xii) a polypeptide comprising the amino acid sequences of SEQ ID NO: 7 or SEQ ID NO: 11, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 22 or SEQ ID NO: 26, respectively,
  • (xiii) a polypeptide comprising the amino acid sequences of SEQ ID NO: 9 or SEQ ID NO: 15, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 24 or SEQ ID NO: 30, respectively,
  • (xiv) a polypeptide comprising the amino acid sequences of SEQ ID NO: 20 or SEQ ID NO: 21, and a DNA encoding the polypeptide, for example, a DNA comprising the nucleotide sequences of SEQ ID NO: 31 or SEQ ID NO: 32, respectively, and
  • (xv) a polypeptide derived from any of the polypeptides described in (i) to (xiv) by comprising the substitution of one or more amino acids in the heavy chain constant region, or a fragment thereof, and a DNA encoding the polypeptide or the fragment thereof.
  • These polypeptides and DNAs can be prepared, as described above, using a gene recombination technique.
  • <Summary of Present Invention>
  • The present invention described above will be summarized below.
  • (1) An antibody which comprises a heavy chain variable region comprising complementarity-determining regions of SEQ ID NOs: 1, 2, and 3 and a light chain variable region comprising complementarity-determining regions of SEQ ID NOs: 4, 5, and 6, and has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof.
  • (2) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
  • (3) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
  • (4) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
  • (5) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 13.
  • (6) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • (7) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
  • (8) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 13.
  • (9) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 14.
  • (10) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
  • (11) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
  • (12) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 9, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
  • (13) The antibody or the fragment thereof according to (1), wherein the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 20, and the light chain variable region comprises the amino acid sequence of SEQ ID NO: 21.
  • (14) The antibody or the fragment thereof according to any of (1) to (13), wherein the antibody is a human antibody, a humanized antibody, a chimeric antibody, a single-chain antibody, or a multispecific antibody.
  • (15) The antibody or the fragment thereof according to any of (1) to (13), wherein the antibody or the fragment is conjugated with an antitumor agent.
  • (16) The antibody according to any of (1) to (15), wherein the antibody comprises the substitution of one or more amino acids in the heavy chain constant region thereof.
  • (17) The antibody according to any of (1) to (16), wherein the antibody is an antibody lacking fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region.
  • (18) An antibody composition comprising an antibody according to (17) and an antibody according to any of (1) to (16) having fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region.
  • (19) A cell producing an antibody according to (17) or an antibody composition according to (18).
  • (20) A pharmaceutical composition for the treatment and/or prevention of a cancer, comprising an antibody or a fragment thereof according to any of (1) to (15), an antibody according to (16) or (17), or an antibody composition according to (18) as an active ingredient.
  • (21) The pharmaceutical composition according to (20), wherein the cancer is breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
  • (22) A combination drug product for the treatment and/or prevention of a cancer, comprising a pharmaceutical composition according to (20) or (21) and a pharmaceutical composition comprising an antitumor agent.
  • (23) A DNA encoding an antibody or a fragment thereof according to any of (1) to (16).
  • (24) A method for treating and/or preventing a cancer, comprising administering an antibody or a fragment thereof according to any of (1) to (16), an antibody according to (17), an antibody composition according to (18), a pharmaceutical composition according to (20) or (21), or a combination drug product according to (22) to a subject.
  • EXAMPLES
  • Hereinafter, the present invention will be described more specifically with reference to Examples. However, the scope of the present invention is not intended to be limited by these specific examples.
  • Example 1 Preparation of Anti-CAPRIN-1 Monoclonal Antibody Using Rabbit
  • 300 μg of human CAPRIN-1 protein prepared in Example 3 of WO2010/016526 was mixed with an equal amount of a Freund's complete adjuvant, and this was used as an antigen solution per rabbit. A mixture with a Freund's incomplete adjuvant was used in the second or later immunization. The antigen solution was intraperitoneally administered to each 12-week-old rabbit, and then, administered 8 times every 2 to 3 weeks to complete immunization. Lymphocytes were obtained from the spleen of each rabbit excised 4 days after the final immunization, and mixed with rabbit myeloma cells 240E-W2 at a ratio of 1:2. A PEG solution (heated to 37° C.) prepared by mixing 200 μL of an RPMI medium containing 10% FBS and 800 μL of PEG1500 was added thereto, and the mixture was left standing for 5 minutes fuse cells. After centrifugation and removal of the supernatant, the cells were suspended in 300 mL of an RPMI medium containing 10% FBS supplemented with HAT solution (HAT selective medium) at a concentration of 2% and inoculated at 100 μL/well to 80 96-well plates. Hybridomas generated by the fusion of the spleen cells and the rabbit myeloma cells were obtained by culturing at 37° C. for 7 days under conditions of 5% CO2.
  • Hybridomas were selected based on the reactivity of antibodies produced by the prepared hybridomas with the CAPRIN-1 protein. A 1 μg/mL CAPRIN-1 protein solution was added at 100 μL/well to the 96-well plates, and the plates were left standing at 4° C. for 18 hours. Each well was washed with PBS-T three times. Then, a 0.5% bovine serum albumin (BSA) solution was added at 400 μL/well, and the plates were left standing at room temperature for 3 hours. The solution was removed, and the wells were washed with 400 μL/well of PBS-T three times. Then, each culture supernatant of the hybridoma obtained above was added at 100 μL/well, and the plates were left standing at room temperature for 2 hours. Each well was washed with PBS-T three times. Then, an HRP-labeled anti-rabbit antibody diluted 5000-fold with PBS was added at 100 μL/well, and the plates were left standing at room temperature for 1 hour. Each well was washed with PBS-T three times. Then, a TMB substrate solution was added at 100 μL/well, and the plates were left standing for 15 to 30 minutes for chromogenic reaction. After the color development, the reaction was terminated by the addition of I N sulfuric acid at 100 μL/well, and the absorbance values were measured at 450 nm and 595 nm using an absorption spectrometer. As a result, a plurality of hybridomas producing antibodies that exhibited a high absorbance values were selected.
  • The selected hybridomas were added at 0.5 cells/well to 96-well plates and cultured. After 1 week, hybridomas that formed single colonies in the wells were observed. The cells in these wells were further cultured, and hybridomas were selected based on the reactivity of antibodies produced by the cloned hybridomas with the CAPRIN-1 protein. As a result of evaluating the reactivity of each antibody with the CAPRIN-1 protein by the same procedure as above, a plurality of hybridoma lines producing rabbit monoclonal antibodies that exhibited reactivity with the CAPRIN-1 protein were obtained.
  • Next, these rabbit monoclonal antibodies that exhibited reactivity with the CAPRIN-1 protein were screened for ones exhibiting reactivity with the surface of human cancer cells on which CAPRIN-1-was expressed. Specifically, 2×105 cells each of a human lung cancer cell line QG56 and a human breast cancer cell line BT-474 (obtained from ATCC) were centrifuged in each 1.5 mL microcentrifuge tube, to which 100 μL of the culture supernatant of each of the hybridomas was then added. The tube was left standing on ice for 1 hour. After washing with PBS, an FITC-labeled anti-rabbit IgG (H+L) antibody or Alexa 488-labeled anti-rabbit IgG (H+L) diluted 100-fold with PBS(−) containing 0.5% FBS (0.5% FBS-PBS(−)) was added thereto, and the tube was left standing on ice for 1 hour. After washing with 0.5% FBS-PBS(−), the cells were suspended in 0.2 μg/mL propidium iodide and 0.5% FBS-PBS(−), and the fluorescence intensity was measured using FACSCalibur™ or FACSVerse™ (Becton. Dickinson and Company). On the other hand, the same procedure as above was carried out using a medium for hybridoma culture, and the resultant was used as a sample for a negative control. As a result, one rabbit anti-CAPRIN-1 monoclonal antibody that exhibited stronger fluorescence intensity than that of the negative control, i.e., strongly reacted with the cell surface of the cancer cells QG56 and BT-474 on expressing CAPRIN-1, was selected.
  • Next, amplification fragments of variable region-encoding genes as to the rabbit anti-CAPRIN-1 monoclonal antibody obtained above were obtained according to the method described in Example 5 of WO2010/016526, and analyzed for their gene sequences and the amino acid sequences encoded thereby. Specifically. mRNA was extracted from the hybridoma producing the rabbit anti-CAPRIN-1 monoclonal antibody, and the genes of the heavy chain variable (VH) region and the light chain variable (VL) region of this antibody were obtained by RT-PCR using primers specific for rabbit variable region sequences. These genes were inserted to cloning vectors, and their respective nucleotide sequences were determined according to a routine method.
  • The resulting rabbit anti-CAPRIN-1 monoclonal antibody was confirmed to have a heavy chain variable region represented by SEQ ID NO: 20, the heavy chain variable region containing CDR1, CDR2, and CDR3 consisting of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and to have a light chain variable region represented by SEQ ID NO: 21, the light chain variable region containing CDR1. CDR2, and CDR3 consisting of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively.
  • Next, the reactivity of the obtained rabbit anti-CAPRIN-1 monoclonal antibody with various human cancer cells was confirmed. The obtained antibody was reacted with human cancer cells confirmed to express the gene of CAPRIN-1, i.e., breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer cells (A-673), Ewing's tumor cells (RD-ES), Hodgkin's lymphoma cells (RPMI1666), mesothelioma cells (NCI-H2452), multiple myeloma cells (IM-9), testicle cancer cells (NT/DI), thyroid cancer cells (TT), or head and neck cancer cells (FaDu), and the fluorescence intensity was evaluated by flow cytometry. 106 cells of each cancer cell line were collected into each 1.5 mL microcentrifuge tube, and the culture supernatant (100 μL) of the hybridoma producing the rabbit anti-CAPRIN-1 monoclonal antibody obtained above was added to each tube and reacted at 4° C. for 1 hour. After washing with 0.5% FBS-PBS(−), an FITC-labeled goat anti-rabbit IgG (H+L) antibody (manufactured by Jackson ImmunoResearch Laboratories, Inc.) diluted 50-fold with 0.5% FBS-PBS(−) was added thereto, and the tube was left standing at 4° C. for 60 minutes. After washing with 0.5% FBS-PBS(−), the cells were suspended in 0.5% FBS-PBS(−) containing 0.2 μg/mL (final concentration) propidium iodide, and the fluorescence intensity was measured using FACSCalibur® or FACSVerse™ (Becton, Dickinson and Company). On the other hand, the same procedure as above was carried out for a negative control using a medium for hybridoma culture, and the resultant prepared was used as a sample for a negative control. As a result, in all of the cancer cells used in the evaluation, the fluorescence intensity when using the culture supernatant of the hybridoma producing the rabbit anti-CAPRIN-1 monoclonal antibody was stronger than that when using the negative control. From these results, the rabbit anti-CAPRIN-1 monoclonal antibody was confirmed to react with CAPRIN-1 on the cancer cell membrane surface of human cancer cells.
  • Example 2 Preparation of Human-Rabbit Chimeric Anti-CAPRIN-1 Monoclonal Antibody
  • The gene for expressing the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 20 of the rabbit anti-CAPRIN-1 monoclonal antibody confirmed in Example 1 and the gene for expressing the light chain variable region represented by SEQ ID NO: 21 thereof were respectively inserted to a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG and a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1. The prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant containing a human-rabbit chimeric anti-CAPRIN-1 antibody (human-rabbit chimeric antibody) was obtained. The obtained culture supernatant containing the chimerized antibody was purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare the chimeric antibody.
  • Example 3 Preparation of Humanized Anti-CAPRIN-1 Monoclonal Antibodies
  • Next, on the basis of information on the amino acid sequences and the nucleotide sequences of CDR1 to CDR3 in the heavy chain variable region of the rabbit anti-CAPRIN-1 monoclonal antibody confirmed in Example 1 and CDR1 to CDR3 in the light chain variable region thereof, a nucleotide sequence was designed so as to be able to express the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 7 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively. This was inserted to a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1. Similarly, a nucleotide sequence was designed so as to be able to express the amino acid sequence of a light chain variable region represented by SEQ ID NO: 11 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively. This was inserted to a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1. These two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant containing humanized antibody #0 consisting of the heavy chain full-length amino acid sequence represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 11 was obtained.
  • Similarly, a culture supernatant containing humanized antibody #1 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 8 in which heavy chain variable region CDR1. CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: I, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the amino acid sequence of the light chain variable region represented by SEQ ID NO: 11, was obtained.
  • Similarly, culture supernatants containing the following humanized antibodies #2 to 10 were further obtained:
  • humanized antibody #2 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 12 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #3 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 13 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #4 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 12;
  • humanized antibody #5 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 13;
  • humanized antibody #6 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively;
  • humanized antibody #7 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 8 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15;
  • humanized antibody #8 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 9 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the light chain full-length amino acid sequence represented by SEQ ID NO: 15.
  • humanized antibody #9 consisting of the amino acid sequence of a heavy chain variable region represented by SEQ ID NO: 10 in which heavy chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 1, SEQ ID NO: 2, and SEQ ID NO: 3, respectively, and the light chain full-length amino acid sequence represented by SEQ ID NO: 14 in which light chain variable region CDR1, CDR2, and CDR3 consisted of the amino acid sequences of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 6, respectively; and
  • humanized antibody #10 consisting of the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 10 and the light chain full-length amino acid sequence represented by SEQ ID NO: 15:
  • The obtained culture supernatants containing the humanized antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare the humanized antibodies.
  • Example 4 Antigen Specificity of Human-Rabbit Chimeric Antibody and Humanized Antibodies #0 to #10 and Reactivity Thereof with Cancer Cells
  • Next, the specific reactivity of the human-rabbit chimeric antibody prepared in Example 2 and the humanized antibodies #0 to #10 prepared in Example 3 with the CAPRIN-1 protein was confirmed by ELISA according to a routine method. Specifically, at first, a PBS solution containing 5 μg/mL CAPRIN-1 protein was added at 100 μL/well to 96-well plates, and the plates were left standing at 4° C. for 18 hours. Each well was washed with PBS-T. Then, a blocking solution consisting of a PBS solution containing 5% skimmed milk was added at 400 μL/well, and the plates were left standing at room temperature for 3 hours.
  • The solution was removed, and each well was washed with PBS-T. Then, a solution containing each of the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 adjusted to 1 μg/mL with PBS containing 0.2% skimmed milk was added at 50 μL/well to each well, and the plates were left standing at room temperature for 1 hour. A well to which of a human IgG antibody confirmed not to react with the CAPRIN-1 protein was added at the same antibody concentration as above, and a well to which no antibody was added were prepared as negative controls together therewith. Each well was washed with PBS-T three times. Then, an HRP-labeled anti-human IgG antibody diluted 3000-fold with PBS containing 0.2% skimmed milk was added at 50 μL/well, and the plates were left standing at room temperature for 1 hour. Each well was washed with PBS-T three times. Then, a TMB substrate solution (manufactured by Thermo Fischer Scientific, Inc.) was added at 100 μl/well, and the plates were left standing for 1 to 30 minutes for chromogenic reaction. After the color development, the reaction was terminated by the addition of 1 N sulfuric acid at 100 μL/well, and the absorbance values were measured at 450 nm and 630 nm using an absorption spectrometer. Further, wells on which the CAPRIN-1 protein was not immobilized (non-immobilized wells) were prepared together therewith, and each antibody was added and assayed similarly. As a result, the absorbance value of the well used as a negative control to which a human IgG antibody confirmed not to react with the CAPRIN-1 protein was added, was as low as that of the well to which no antibody was added, whereas the wells respectively to which the respective human-rabbit chimeric antibody and the humanized antibodies #0 to #10 exhibited equivalently high absorbance values. The human-rabbit chimeric antibody and the humanized antibodies #0 to #10 in the wells on which the CAPRIN-1 protein was not immobilized merely exhibited an absorbance value equivalent to that of the negative control. From these results, the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 were confirmed to specifically react with the CAPRIN-1 protein.
  • Next, the reactivity of the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 specifically reacting with the CAPRIN-1 protein, with various human cancer cells and mouse cancer cells was confirmed. The purified human-rabbit chimeric antibody and humanized antibodies #0 to #10 were each reacted with human cancer cells confirmed to express the gene of CAPRIN-1, i.e., breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer cells (A-673), Ewing's tumor cells (RD-ES), Hodgkin's lymphoma cells (RPM11666), mesothelioma cells (NCI-H2452), multiple myeloma cells (IM-9), testicle cancer cells (NT/D I), thyroid cancer cells (TT), or head and neck cancer cells (FaDu), and the fluorescence intensity was evaluated by flow cytometry. Specifically, 5×10′ cells of each cancer cell line were collected into each 1.5 mL microcentrifuge tube, and each of the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 was added at 50 μg/mL (final concentration) to each tube and reacted at 4° C. for 1 hour. After washing with 0.5% FBS-PBS(−) twice, an Alexa 488-labeled goat anti-human IgG (H+L) antibody (manufactured by Life Technologies Corp.) diluted 100-fold with 0.5% FBS-PBS(−) was added thereto, and the tube was left standing at 4° C. for 60 minutes. After washing with 0.5% FBS-PBS(−), the cells were suspended in 0.5% FBS-PBS(−) containing 0.2 μg/mL (final concentration) propidium iodide, and the fluorescence intensity was measured using FACSCalibur™ or FACSVerse™ (Becton, Dickinson and Company). On the other hand, the same procedure as above was carried out using a medium for hybridoma culture, and the resultant prepared was used for a negative control. As a result, in all of the cancer cells used in the evaluation, the fluorescence intensity from the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 was stronger than that in the case of using the negative control. From these results, the human-rabbit chimeric antibody and the humanized antibodies #0 to #10 were confirmed to react with the CAPRIN-1 protein expressed on the membrane surface of human cancer cells.
  • Example 5 Antitumor Activity of Human-Rabbit Chimeric Antibody and Humanized Antibodies #0 to #10 Against Various Human Cancer Cells
  • Next, the human-rabbit chimeric antibody prepared in Example 2 and the humanized antibodies #0 to #10 prepared in Example 3 were evaluated for their antitumor effects on various human cancer cells on the basis of ADCC activity.
  • The following anti-CAPRIN-1 antibodies were used as comparative antibodies for the human-rabbit chimeric antibody and the humanized antibodies #0 to #10:
  • antibodies described in WO2010/016526, i.e., comparative antibody 1 having a heavy chain variable region of SEQ ID NO: 26 and a light chain variable region of SEQ ID NO: 27 in this literature, comparative antibody 2 having a heavy chain variable region of SEQ ID NO: 28 and a light chain variable region of SEQ ID NO: 29 therein, comparative antibody 3 having a heavy chain variable region of SEQ ID NO: 30 and a light chain variable region of SEQ ID NO: 31 therein, comparative antibody 4 having a heavy chain variable region of SEQ ID NO: 32 and a light chain variable region of SEQ ID NO: 33 therein, comparative antibody 5 having a heavy chain variable region of SEQ ID NO: 34 and a light chain variable region of SEQ ID NO: 35 therein, comparative antibody 6 having a heavy chain variable region of SEQ ID NO: 36 and a light chain variable region of SEQ ID NO: 37 therein, comparative antibody 7 having a heavy chain variable region of SEQ ID NO: 38 and a light chain variable region of SEQ ID NO: 39 therein, comparative antibody 8 having a heavy chain variable region of SEQ ID NO: 40 and a light chain variable region of SEQ ID NO: 41 therein, comparative antibody 9 having a heavy chain variable region of SEQ ID NO: 42 and a light chain variable region of SEQ ID NO: 43 therein, comparative antibody 10 having a heavy chain variable region of SEQ ID NO: 44 and a light chain variable region of SEQ ID NO: 45 therein, and comparative antibody 11 having a heavy chain variable region of SEQ ID NO: 46 and a light chain variable region of SEQ ID NO: 47 therein;
  • antibodies described in WO2011/096517, i.e., comparative antibody 12 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 47 in this literature, and comparative antibody 13 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 53 therein:
  • antibodies described in WO2011/096528, i.e., comparative antibody 14 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 47 in this literature, comparative antibody 15 having a heavy chain variable region of SEQ ID NO: 51 and a light chain variable region of SEQ ID NO: 55 therein, comparative antibody 16 having a heavy chain variable region of SEQ ID NO: 59 and a light chain variable region of SEQ ID NO: 63 therein, comparative antibody 17 having a heavy chain variable region of SEQ ID NO: 76 and a light chain variable region of SEQ ID NO: 80 therein, comparative antibody 18 having a heavy chain variable region of SEQ ID NO: 84 and a light chain variable region of SEQ ID NO: 88 therein, and comparative antibody 19 having a heavy chain variable region of SEQ ID NO: 92 and a light chain variable region of SEQ ID NO: 96 therein:
  • an antibody described in WO2011/096519, i.e., comparative antibody 20 having a heavy chain variable region of SEQ ID NO: 42 and a light chain variable region of SEQ ID NO: 46 in this literature;
  • antibodies described in WO2011/096533, i.e., comparative antibody 21 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 51 in this literature, comparative antibody 22 having a heavy chain variable region of SEQ ID NO: 47 and a light chain variable region of SEQ ID NO: 51 therein, and comparative antibody 23 having a heavy chain variable region of SEQ ID NO: 63 and a light chain variable region of SEQ ID NO: 67 therein;
  • antibodies described in WO2011/096534, i.e., comparative antibody 24 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 47 in this literature, comparative antibody 25 having a heavy chain variable region of SEQ ID NO: 43 and a light chain variable region of SEQ ID NO: 51 therein, and comparative antibody 26 having a heavy chain variable region of SEQ ID NO: 63 and a light chain variable region of SEQ ID NO: 67 therein;
  • antibodies described in WO2013/018894, i.e., comparative antibody 27 having a heavy chain variable region of SEQ ID NO: 9 and a light chain variable region of SEQ ID NO: 13 in this literature, comparative antibody 28 having a heavy chain variable region of SEQ ID NO: 19 and a light chain variable region of SEQ ID NO: 23 therein, comparative antibody 29 having a heavy chain variable region of SEQ ID NO: 9 and a light chain variable region of SEQ ID NO: 53 therein, comparative antibody 30 having a heavy chain variable region of SEQ ID NO: 58 and a light chain variable region of SEQ ID NO: 62 therein, comparative antibody 31 having a heavy chain variable region of SEQ ID NO: 63 and a light chain variable region of SEQ ID NO: 65 therein, comparative antibody 32 having a heavy chain variable region of SEQ ID NO: 69 and a light chain variable region of SEQ ID NO: 73 therein, and comparative antibody 33 having a heavy chain variable region of SEQ ID NO: 77 and a light chain variable region of SEQ ID NO: 81 therein:
  • an antibody described in WO2013/018892, i.e., comparative antibody 34 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2013/018891, i.e., comparative antibody 35 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2013/018889, i.e., comparative antibody 36 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2010/018883, i.e., comparative antibody 37 having a heavy chain variable region of SEQ ID NO: 8 and a light chain variable region of SEQ ID NO: 12 in this literature;
  • an antibody described in WO2013/125636, i.e., comparative antibody 38 having a heavy chain variable region of SEQ ID NO: 6 and a light chain variable region of SEQ ID NO: 7 in this literature:
  • antibodies described in WO2013/125654, i.e., comparative antibody 39 having a heavy chain variable region of SEQ ID NO: 52 and a light chain variable region of SEQ ID NO: 54 in this literature, comparative antibody 40 having a heavy chain variable region of SEQ ID NO: 21 and a light chain variable region of SEQ ID NO: 23 therein, comparative antibody 41 having a heavy chain variable region of SEQ ID NO: 25 and a light chain variable region of SEQ ID NO: 23 therein, comparative antibody 42 having a heavy chain variable region of SEQ ID NO: 16 and a light chain variable region of SEQ ID NO: 18 therein, comparative antibody 43 having a heavy chain variable region of SEQ ID NO: 29 and a light chain variable region of SEQ ID NO: 33 therein, comparative antibody 44 having a heavy chain variable region of SEQ ID NO: 39 and a light chain variable region of SEQ ID NO: 43 therein, and comparative antibody 45 having a heavy chain variable region of SEQ ID NO: 49 and a light chain variable region of SEQ ID NO: 43 therein;
  • an antibody described in WO2013/125630, i.e., comparative antibody 46 having a heavy chain variable region of SEQ ID NO: 11 and a light chain variable region of SEQ ID NO: 15 in this literature; and
  • antibodies described in WO2013/125640, i.e., comparative antibody 47 having a heavy chain variable region of SEQ ID NO: 11 and a light chain variable region of SEQ ID NO: 15 in this literature, and comparative antibody 48 having a heavy chain variable region of SEQ ID NO: 21 and a light chain variable region of SEQ ID NO: 25 therein.
  • The aforementioned compared antibodies (comparative antibodies 1 to 48) were each prepared as follows: the gene for expressing the amino acid sequence of the heavy chain variable region and the gene for expressing the light chain variable region were respectively inserted to a vector pcDNA4/myc-His for expression in mammalian cells (manufactured by Life Technologies Corp.) having a gene insert of the heavy chain constant region of human IgG1 and a vector pcDNA3.1/myc-His for expression in mammalian cells (manufactured by Life Technologies Corp.) having a gene insert of the light chain constant region of human IgG1; the prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method; the obtained human-chimerized or humanized antibody was purified using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.); the buffer was replaced with PBS(−); and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.).
  • A well to which an isotype control antibody was added, a well to which no antibody was added, and a well to which an antibody reacting with the CAPRIN-1 protein but exhibiting no reactivity with the surface of human cancer cells, on which CAPRIN-1 was expressed, was added were prepared as negative controls. Each antibody was added at 5 μg/mL (final concentration) to V-bottomed 96-well plates.
  • Human NK cells separated from human peripheral blood mononuclear cells by using a routine method were used as effector cells. The human peripheral blood mononuclear cells were separated using a specific gravity separation solution Histopaque for peripheral blood mononuclear cell separation (Sigma-Aldrich Corp.), and reacted with antibodies (anti-human CD3 antibody, anti-human CD20 antibody, anti-human CD19 antibody, anti-human CD11c antibody, anti-HLA-DR antibody (BD Pharmingen)) labeled with an FITC fluorescent dye. A cell population containing NK cells that were not stained with these antibodies was separated using a cell sorter (FACS Vantage SE (Becton, Dickinson and Company)). Alternatively, a cell population separated using a human NK cell separation kit (manufactured by Miltenyi Biotec K.K.) was used. The V-bottomed 96-well plates to which each of the antibodies was added and the human NK cells were added at 0.4 to 2.0×105 cells/well were prepared.
  • Breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer (A-673), Ewing's tumor (RD-ES), Hodgkin's lymphoma (RPM11666), mesothelioma (NCI-H2452), multiple myeloma (IM-9), testicle cancer (NT/D1), thyroid cancer (TT), and head and neck cancer (FaDu) were used as target cells. 106 cells of each human cancer cell line mentioned above were collected into each 50 mL centrifugal tube. 100 μCi of chromium 51 (manufactured by PerkinElmer, Inc.) was added thereto, and the tube was incubated at 37° C. for 1 hour. Then, the cells were washed with an RPMI1640 medium containing 10% FBS three times, added at 2×103 cells/well to the 96-well V-bottomed plates to which the effector cells and each antibody were added as described above, and reacted at 37° C. for 4 hours under conditions of 5% CO2. After the reaction, 50 μL of a culture supernatant containing chromium 51 released into the culture supernatant from damaged cancer cells was recovered from each well, added to LumaPlate-96 (manufactured by PerkinElmer, Inc.) with the bottom of each well coated with a solid scintillator, and dried. The amount of chromium 51 released into the culture supernatant from damaged cancer cells was measured to calculate the antitumor effects of the anti-CAPRIN-1 antibodies on the cancer cells.
  • As a result, for the breast cancer cells (BT-474), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 54% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 50% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 46% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 25% or lower activity, and all of the negative control groups exhibited 10% or lower activity.
  • For the breast cancer cells (MDA-MB-361), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 52% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 25% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the colorectal cancer cells (HT-29), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 43% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 35% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 3% or lower activity.
  • For the lung cancer cells (QG56), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 46% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 42% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 38% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 22% or lower activity, and all of the negative control groups exhibited 10%0 or lower activity.
  • For the stomach cancer cells (NCI-N87), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 45% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 38% or higher activity, and the antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 34% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 8% or lower activity.
  • For the uterine cancer cells (HEC-1-A), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 52% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the prostate cancer cells (22Rv1), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 49% or higher antitumor effect, the humanized antibody #3, the antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 45% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 38% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 12% or lower activity.
  • For the pancreatic cancer cells (Panc10.5), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 24% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 2% or lower activity.
  • For the liver cancer cells (Hep3B), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 28% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 25% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 21% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 12% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the ovary cancer cells (SKOV3), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 31% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 27% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the kidney cancer cells (Caki-2), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 37% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 26% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the brain tumor cells (U-87MG), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 36% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 29% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 24% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the bladder cancer cells (T24), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 36% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the bladder cancer cells (HT-1376), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 45% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 28% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 7% or lower activity.
  • For the esophagus cancer cells (OE33), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 33% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the leukemia cells (OCI-AML5), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 20% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 18% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the lymphoma cells (Ramos), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 20% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 18% or higher activity, and the antibody #9, the antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the gallbladder cancer cells (TGBC14TKB), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 25% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the fibrosarcoma cells (HT-1080), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 30% or higher antitumor effect, the humanized antibody #3, the antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 25% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 20% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the melanoma (G-361), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 25% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 21% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 15% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 8% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the adrenal cortex cancer cells (A-673), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 50% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 46% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 40% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 8% or lower activity.
  • For the Ewing's tumor cells (RD-ES), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 48% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 31% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the Hodgkin's lymphoma cells (RPM11666), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 40% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 36% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the mesothelioma cells (NCI-H2452), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 39% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 31% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the multiple myeloma cells (IM-9), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 35% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the antibody #2, and the humanized antibody #5 exhibited 30% or higher activity, and the humanized antibody #9, the antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 27% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 10%0 or lower activity, and all of the negative control groups exhibited 6% or lower activity.
  • For the testicle cancer cells (NT/D1), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 37% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 30% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 25% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 11% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the thyroid cancer cells (TT), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 42% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 35% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the humanized antibody #8 exhibited 30% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 15% or lower activity, and all of the negative control groups exhibited 5% or lower activity.
  • For the head and neck cancer cells (FaDu), the humanized antibody #7, the humanized antibody #10, and the humanized antibody #6 exhibited 50% or higher antitumor effect, the humanized antibody #3, the humanized antibody #4, the humanized antibody #2, and the humanized antibody #5 exhibited 40% or higher activity, and the humanized antibody #9, the humanized antibody #1, the humanized antibody #0, the human-rabbit chimeric antibody, and the antibody #8 exhibited 35% or higher activity. By contrast, all of the comparative antibodies 1 to 48 exhibited 20% or lower activity, and all of the negative control groups exhibited 8% or lower activity.
  • These results demonstrated that the humanized antibodies #0 to #10 and the human-rabbit chimeric antibody exhibit a significantly stronger antitumor effect on breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer than that of the comparative antibodies.
  • Moreover, the humanized antibodies #0 to #10 and the human-rabbit chimeric antibody exhibited significantly stronger antitumor activity against breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer as described above than that of all of the antibodies against CAPRIN-1 described in Examples of WO2010/016526, WO2011/096517, WO2011/096528, WO2011/096519. WO2011/096533. WO2011/096534. WO2011/096535. WO2013/018886. WO20131018894, WO2013/018892, WO2013/018891, WO2013/018889, WO2013/018883, WO2013/125636, WO2013/125654, WO2013/125630, WO2013/125640, WO2013/147169, and WO2013/147176.
  • The antitumor effect was shown as cytotoxic activity against the cancer cell lines that was determined by mixing each antibody against CAPRIN-1, effector cells, and chromium 51-incorporated target cells, culturing the cells for 4 hours, and measuring the amount of chromium 51 released into the medium after the cultivation, followed by calculating according to the following formula*:

  • Cytotoxic activity (%)=(Amount of chromium 51 released from the target cells when the antibody against CAPRIN-1 and effector cells are added−Amount of chromium 51 naturally released from the target cells)/(Amount of chromium 51 released from the target cells to which 1 N hydrochloric acid is added−Amount of chromium 51 naturally released from the target cells)×100.  *Formula:
  • Example 6-1 Preparation of Humanized Anti-CAPRIN-1 Monoclonal Antibodies in which Amino Acid in Heavy Chain Constant Region was Substituted
  • Anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 33 in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 was substituted (hereinafter, referred to as engineered I-type anti-CAPRIN-1 antibodies) were prepared. A DNA encoding the amino acid sequence of a heavy chain having the heavy chain constant region mentioned above and a heavy chain variable region represented by SEQ ID NO: 7 was synthesized, and this was inserted to a vector for expression in mammalian cells according to a routine method. Furthermore, a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was inserted to a vector for expression in mammalian cells having an insert of a gene encoding the light chain constant region of human IgG to prepare a recombinant expression vector. The prepared two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant of engineered I-type anti-CAPRIN-1 antibody #0 of the humanized antibody #0 was obtained. Culture supernatants containing engineered I-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3 were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively. The obtained culture supernatants containing the engineered I-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare the engineered I-type anti-CAPRIN-1 antibodies.
  • Next, anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 34, in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 was substituted (hereinafter, referred to as engineered II-type anti-CAPRIN-1 antibodies), were prepared. A DNA encoding the amino acid sequence of a heavy chain having the heavy chain constant region mentioned above and a heavy chain variable region represented by SEQ ID NO: 7 was synthesized, and this was inserted to a vector for expression in mammalian cells according to a routine method. Further, as prepared above, a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was inserted to a vector for expression in mammalian cells having an insert of a gene encoding the light chain constant region of human IgG1 to prepare a recombinant expression vector. These two recombinant expression vectors were transferred to mammalian cells according to a routine method, and a culture supernatant of engineered II-type anti-CAPRIN-1 antibody #0 was obtained. Culture supernatants containing engineered II-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3, were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively. The obtained culture supernatants containing the engineered II-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare the engineered II-type anti-CAPRIN-1 antibodies.
  • Example 6-2 Preparation of Anti-CAPRIN-1 Antibodies Having Sugar Chain with No Fucose Added to N-Acetylglucosamine at Sugar Chain Reducing End Among all N-Glycoside-Linked Sugar Chains Attached to Heavy Chain Constant Region
  • Next, anti-CAPRIN-1 antibodies having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 obtained in Example 3 (hereinafter, referred to as engineered III-type anti-CAPRIN-1 antibodies) were obtained by the following method: a neomycin resistance gene-containing mammalian cell expression vector harboring a gene of GDP-6-deoxy-D-lyxo-4-hexulose reductase (RMD), which is an enzyme that does not catalyze the reaction of converting GDP-6-deoxy-D-lyxo-4-hexulose to GDP-L-fucose, was transferred to mammalian cell line CHO cells according to a routine method using a transfection reagent FreeStyle™ MAX Reagent (Life Technologies Corp.). The transfected CHO cells were cultured in a medium containing G-418 to prepare a stable pool of CHO cells expressing RMD. From this stable pool, 7 CHO cells constitutively expressing RMD were cloned by the limiting dilution method. The RMD gene expression level in each of the cloned 7 RMD-CHO cells was evaluated three times every other week by quantitative PCR to select CHO cells constitutively and stably expressing the RMD gene (RMD-CHO cells). In the same way as in Example 3, the gene encoding the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the gene encoding the amino acids of the light chain variable region represented by SEQ ID NO: 11 were respectively transferred through a vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1 and a vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1 to the RMD-CHO cells constitutively expressing RMD according to a routine method, and a culture supernatant containing engineered III-type anti-CAPRIN-1 antibody #0 was obtained. Culture supernatants containing engineered III-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3, were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively. The obtained culture supernatants containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare an antibody composition containing the engineered III-type anti-CAPRIN-1 antibody #0. Similarly, antibody purified products containing engineered III-type anti-CAPRIN-1 antibodies #1 to #10 were obtained.
  • The proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of these purified antibody compositions was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • In the same way as in Example 3, the gene encoding the amino acid sequence of the heavy chain variable region represented by SEQ ID NO: 7 and the gene encoding the amino acids of the light chain variable region represented by SEQ ID NO: 11 were respectively transferred through a hygromycin resistance gene-containing vector for expression in mammalian cells having a gene insert of the heavy chain constant region of human IgG1 and a hygromycin resistance gene-containing vector for expression in mammalian cells having a gene insert of the light chain constant region of human IgG1 to the RMD-CHO cells according to a routine method, and the resulting cells were cultured in a medium containing hygromycin B to prepare a stable pool expressing engineered III-type anti-CAPRIN-1 antibody #0. From this stable pool, cells constitutively and stably expressing the engineered III-type anti-CAPRIN-1 antibody #0 were prepared by the limiting dilution method. The proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of the purified antibody compositions comprising the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 produced by the respective cells was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • Example 6-3 Preparation of Anti-CAPRIN-1 Antibodies Having Amino Acid Substitution in Heavy Chain Constant Region and Having Sugar Chain with No Fucose Added to N-Acetylglucosamine at Sugar Chain Reducing End Among all N-Glycoside-Linked Sugar Chains Attached to Heavy Chain Constant Region
  • Next, anti-CAPRIN-1 antibodies having a heavy chain constant region described in SEQ ID NO: 34 in which a portion of amino acids in the heavy chain constant region of the humanized antibody #0, #1, #2, #3, #4, #5, #6, #7, #8, #9, or #10 described in Example 3 was substituted, and having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region of the antibody (hereinafter, referred to as engineered N-type anti-CAPRIN-1 antibodies) were prepared. A DNA encoding the amino acid sequence of a heavy chain having the variant heavy chain constant region prepared in Example 6-1 and the heavy chain variable region of human IgG1 represented by SEQ ID NO: 7 was synthesized and inserted to a vector for expression in mammalian cells according to a routine method, while a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was synthesized and inserted to a vector for expression in mammalian cells having an insert of a gene encoding the amino acids of the light chain constant region of human IgG1, and the resulting vectors were transferred to the RMD-CHO cells constitutively expressing GDP-6-deoxy-D-lyxo-4-hexulose reductase (RMD) prepared in Example 6-2. A culture supernatant of engineered N-type anti-CAPRIN-1 antibody #0 of the humanized antibody #0 was obtained. Culture supernatants containing engineered N-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 were further obtained in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10, respectively, described in Example 3. The obtained culture supernatants containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 were each purified according to a routine method using Hitrap Protein A Sepharose FF (manufactured by GE Healthcare Japan Corp.). The buffer was replaced with PBS(−), and the resultant was filtered through a 0.22 μm filter (manufactured by Merck Millipore Corp.) to prepare an antibody composition containing the engineered IV-type anti-CAPRIN-1 antibody #0. Similarly, antibody purified products containing engineered IV-type anti-CAPRIN-1 antibodies #1 to #10 were obtained. The proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of these purified antibody compositions was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • A DNA encoding the amino acid sequence of a heavy chain having the modified heavy chain constant region prepared in Example 6-1 and the heavy chain variable region of human IgG1 represented by SEQ ID NO: 7 was synthesized and inserted to a hygromycin resistance gene-containing vector for expression in mammalian cells according to a routine method, while a DNA encoding the amino acids of a light chain variable region represented by SEQ ID NO: 11 was synthesized and inserted to a hygromycin resistance gene-containing vector for expression in mammalian cells having an insert of a gene encoding the amino acids of the light chain constant region of human IgG1, and the resulting vectors were transferred to the cells. The cells were cultured in a medium containing hygromycin B to prepare a stable pool expressing engineered IV-type anti-CAPRIN-1 antibody #0. From this stable pool, cells constitutively and stably expressing the engineered IV-type anti-CAPRIN-1 antibody #0 were prepared by the limiting dilution method. Cells constitutively and stably expressing engineered IV-type anti-CAPRIN-1 antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 were further prepared in the same way as above as to the humanized antibodies #1, #2, #3, #4, #5, #6, #7, #8, #9, and #10 described in Example 3, respectively. The proportion of the anti-CAPRIN-1 antibody having a sugar chain with no fucose added to N-acetylglucosamine at the sugar chain reducing end among all N-glycoside-linked sugar chains attached to the heavy chain constant region, contained in each of the purified antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 produced by the respective cells was evaluated using LabChip® GXII (PerkinElmer, Inc.) and consequently, was 80% or higher in all cases.
  • Example 7 Antigen Specificity of Engineered-Type Anti-CAPRIN-1 Antibodies and Reactivity Thereof with Cancer Cells
  • The specific reactivity of the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 (hereinafter, referred to as engineered-type anti-CAPRIN-1 antibodies) prepared in Examples 6-1 to 6-3 with the CAPRIN-1 protein was confirmed in the same way as in Example 4. As a result, the absorbance value of the well to which a human IgG antibody confirmed not to react with the CAPRIN-1 protein was added, used as a negative control, was as low as that of the well to which no antibody was added, whereas all of the wells respectively to which the engineered-type anti-CAPRIN-1 antibodies were added exhibited equivalently high absorbance values. All of the engineered-type anti-CAPRIN-1 antibodies in the wells on which the CAPRIN-1 protein was not immobilized merely exhibited an absorbance value equivalent to that of the negative control. From these results, all of the engineered-type anti-CAPRIN-1 antibodies were confirmed to specifically react with the CAPRIN-1 protein.
  • Next, the reactivity of each engineered-type anti-CAPRIN-1 antibody with various human cancer cells was confirmed in the same way as in Example 4. Breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer cells (A-673), Ewing's tumor cells (RD-ES), Hodgkin's lymphoma cells (RPMI11666), mesothelioma cells (NCI-H2452), multiple myeloma cells (IM-9), testicle cancer cells (NT/D1), thyroid cancer cells (TT), and head and neck cancer cells (FaDu) were used in this evaluation. As a result, the fluorescence intensity from each engineered-type anti-CAPRIN-1 antibody was stronger in all of the cancer cells used in the evaluation than that in the case of using the negative control. From these results, all of the engineered-type anti-CAPRIN-1 antibodies were confirmed to specifically react with the CAPRIN-1 protein present on the membrane surface of human cancer cells.
  • Example 8 Antitumor Activity of Engineered-Type Anti-CAPRIN-1 Antibody Against Various Human Cancer Cells
  • The engineered-type anti-CAPRIN-1 antibodies (the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10) prepared in Examples 6-1 to 6-3 were evaluated for their antitumor effects on various human cancer cells on the basis of ADCC activity in the same way as in Example 5. A well to which an isotype control antibody was added, a well to which no antibody was added, and a well to which an antibody reacting with the CAPRIN-1 protein but exhibiting no reactivity with the surface of human cancer cells, on which CAPRIN-1 was expressed, was added were prepared as negative controls. The humanized antibodies #0 to #10, which were the corresponding unengineered anti-CAPRIN-1 antibodies, were used as comparative antibodies. Each antibody was added at 0.01 to 1 μg/mL (final concentration) to V-bottomed 96-well plates.
  • Breast cancer cells (BT-474 and MDA-MB-361), colorectal cancer cells (HT-29), lung cancer cells (QG56), stomach cancer cells (NCI-N87), uterine cancer cells (HEC-1-A), prostate cancer cells (22Rv1), pancreatic cancer cells (Panc10.5), liver cancer cells (Hep3B), ovary cancer cells (SKOV3), kidney cancer cells (Caki-2), brain tumor cells (U-87MG), bladder cancer cells (T24 and HT-1376), esophagus cancer cells (OE33), leukemia cells (OCI-AML5), lymphoma cells (Ramos), gallbladder cancer cells (TGBC14TKB), fibrosarcoma cells (HT-1080), melanoma cells (G-361), adrenal cortex cancer cells (A-673), Ewing's tumor cells (RD-ES), Hodgkin's lymphoma cells (RPMI1666), mesothelioma cells (NCI-H2452), multiple myeloma cells (IM-9), testicle cancer cells (NT/D1), thyroid cancer cells (Ti), and head and neck cancer cells (FaDu) were used as target cells. 106 cells of each human cancer cell line mentioned above were collected into each 50 mL centrifugal tube. 100 μCi of chromium 51 (manufactured by PerkinElmer, Inc.) was added thereto, and the tube was incubated at 37° C. for 1 hour. Then, the cells were washed with an RPMI1640 medium containing 10% FBS three times, added at 2×103 cells/well to the 96-well V-bottomed plates to which each of the antibodies was added, and reacted.
  • Human NK cells separated from human peripheral blood mononuclear cells by using a routine method were used as effector cells. The human NK cells added at 0.4 to 2.0×105 cells/well to the V-bottomed 96-well plates to which each of the antibodies and the target cells were added and reacted were prepared and reacted at 37° C. for 4 hours under conditions of 5% CO2. After the reaction, 50 μL of a culture supernatant containing chromium 51 released from damaged cancer cells was recovered from each well. The amount of chromium 51 released into the culture supernatant from damaged cancer cells was measured in the same way as in Example 5 to calculate the antitumor effects of the anti-CAPRIN-1 antibodies on the cancer cells.
  • As a result, for the breast cancer cells (BT-474), the engineered-type anti-CAPRIN-1 antibodies, i.e., the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 each exhibited a stronger antitumor effect than that of the negative controls. The engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 exhibited the same level of the antitumor effect as that exhibited by the corresponding unengineered antibodies (humanized antibodies #0 to #10) used as comparative antibodies were approximately 1/13 to 1/20 concentrations of the unengineered antibody concentrations, respectively. In order for the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 to achieve the same level of the antitumor effect as that of the corresponding unengineered antibodies, the concentrations were approximately 1/150 of the unengineered antibody concentrations, respectively. These results demonstrated that the engineered-type anti-CAPRIN-1 antibodies (the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered III-type anti-CAPRIN-1 antibodies #0 to #10 and the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10) exhibit improved antitumor activity as compared with the corresponding unengineered antibodies. These results also demonstrated that the respective antibody compositions containing the engineered IV-type anti-CAPRIN-1 antibodies #0 to #10 produce a stronger antitumor effect than that of the engineered I-type anti-CAPRIN-1 antibodies #0 to #10, the engineered II-type anti-CAPRIN-1 antibodies #0 to #10, and the respective antibody compositions containing the engineered 11I-type anti-CAPRIN-1 antibodies #0 to #10.
  • In addition, a similar stronger antitumor effect was also obtained for the breast cancer cells (MDA-MB-361), the colorectal cancer cells (HT-29), the lung cancer cells (QG56), the stomach cancer cells (NCI-N87), the uterine cancer cells (HEC-1-A), the prostate cancer cells (22Rv1), the pancreatic cancer cells (Panc10.5), the liver cancer cells (Hep3B), the ovary cancer cells (SKOV3), the kidney cancer cells (Caki-2), the brain tumor cells (U-87MG), the bladder cancer cells (T24 and HT-1376), the esophagus cancer cells (OE33), the leukemia cells (OCI-AML5), the lymphoma cells (Ramos), the gallbladder cancer cells (TGBC14TKB), the fibrosarcoma cells (HT-1080), the melanoma cells (G-361), the adrenal cortex cancer cells (A-673), the Ewing's tumor cells (RD-ES), the Hodgkin's lymphoma cells (RPMII666), the mesothelioma cells (NCI-H2452), the multiple myeloma cells (IM-9), the testicle cancer cells (NT/D1), the thyroid cancer cells (TT), and the head and neck cancer cells (FaDu) used in the evaluation.
  • INDUSTRIAL APPLICABILITY
  • The antibody of the present invention is useful for the treatment and/or prevention of a cancer.
  • All publications, patents, and patent applications cited herein are incorporated herein by reference in their entirety.

Claims (24)

1. An antibody which comprises
a heavy chain variable region comprising complementarity-determining regions of SEQ ID NOs: 1, 2, and 3 and
a light chain variable region comprising complementarity-determining regions of SEQ ID NOs: 4, 5, and 6, and
has immunological reactivity with a CAPRIN-1 protein, or a fragment thereof.
2. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
3. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
4. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
5. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 13.
6. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
7. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 12.
8. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 13.
9. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 10, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 14.
10. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 8, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
11. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 7, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 11.
12. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 9, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 15.
13. The antibody or the fragment thereof according to claim 1,
wherein
the heavy chain variable region comprises the amino acid sequence of SEQ ID NO: 20, and
the light chain variable region comprises the amino acid sequence of SEQ ID NO: 21.
14. The antibody or the fragment thereof according to claim 1, wherein the antibody is a human antibody, a humanized antibody, a chimeric antibody, a single-chain antibody, or a multispecific antibody.
15. The antibody or the fragment thereof according to claim 1, wherein the antibody or the fragment is conjugated with an antitumor agent.
16. The antibody according to claim 1, wherein the antibody comprises the substitution of one or more amino acids in the heavy chain constant region thereof.
17. The antibody according to claim 1, wherein the antibody is an antibody lacking fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region.
18. An antibody composition comprising:
an antibody wherein the antibody is an antibody lacking fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region, and
an antibody according to claim 1 having fucose added to N-acetylglucosamine at the sugar chain reducing end of a N-glycoside-linked sugar chain attached to the heavy chain constant region.
19. A cell producing an antibody according to claim 17.
20. A pharmaceutical composition for the treatment and/or prevention of a cancer, comprising an antibody or a fragment thereof according to claim 1 as an active ingredient.
21. The pharmaceutical composition according to claim 20, wherein the cancer is breast cancer, kidney cancer, pancreatic cancer, colorectal cancer, lung cancer, brain tumor, stomach cancer, uterine cancer, ovary cancer, prostate cancer, bladder cancer, esophagus cancer, leukemia, lymphoma, liver cancer, gallbladder cancer, sarcoma, mastocytoma, melanoma, adrenal cortex cancer, Ewing's tumor, Hodgkin's lymphoma, mesothelioma, multiple myeloma, testicle cancer, thyroid cancer, or head and neck cancer.
22. A combination drug product for the treatment and/or prevention of a cancer, comprising a pharmaceutical composition according to claim 20 and a pharmaceutical composition comprising an antitumor agent.
23. A DNA encoding an antibody or a fragment thereof according to claim 1.
24. A method for treating and/or preventing a cancer, comprising administering an antibody or a fragment thereof according to claim 1 to a subject.
US14/910,878 2013-08-09 2014-08-08 Pharmaceutical composition for treatment and/or prevention of cancer Active US9862774B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2013-166164 2013-08-09
JP2013166164 2013-08-09
PCT/JP2014/071094 WO2015020212A1 (en) 2013-08-09 2014-08-08 Pharmaceutical composition for treatment and/or prevention of cancer

Publications (2)

Publication Number Publication Date
US20160297889A1 true US20160297889A1 (en) 2016-10-13
US9862774B2 US9862774B2 (en) 2018-01-09

Family

ID=52461536

Family Applications (1)

Application Number Title Priority Date Filing Date
US14/910,878 Active US9862774B2 (en) 2013-08-09 2014-08-08 Pharmaceutical composition for treatment and/or prevention of cancer

Country Status (17)

Country Link
US (1) US9862774B2 (en)
EP (1) EP3031826B1 (en)
JP (1) JP6447130B2 (en)
KR (1) KR102255616B1 (en)
CN (1) CN105452294B (en)
AU (1) AU2014303375B2 (en)
BR (1) BR112016001753B1 (en)
CA (1) CA2918989C (en)
DK (1) DK3031826T3 (en)
ES (1) ES2704909T3 (en)
HU (1) HUE042081T2 (en)
MX (1) MX360671B (en)
PL (1) PL3031826T3 (en)
PT (1) PT3031826T (en)
RU (1) RU2678138C2 (en)
TR (1) TR201819812T4 (en)
WO (1) WO2015020212A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160208014A1 (en) * 2008-08-05 2016-07-21 Toray Industries, Inc. Pharmaceutical composition for treatment and prevention of cancers
US9796775B2 (en) 2011-08-04 2017-10-24 Toray Industries, Inc. Method for detecting pancreatic cancer
US9862774B2 (en) 2013-08-09 2018-01-09 Toray Industries, Inc. Pharmaceutical composition for treatment and/or prevention of cancer
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11137401B2 (en) 2008-08-05 2021-10-05 Toray Industries, Inc. Method for detecting cancer using CAPRIN-1 as a marker
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109890417A (en) 2016-10-28 2019-06-14 东丽株式会社 The treatment of cancer and/or prophylactic compositions
CA3095719A1 (en) * 2018-03-30 2019-10-03 Toray Industries, Inc. Pharmaceutical composition for treatment and/or prevention of cancer
KR20220034784A (en) 2019-06-13 2022-03-18 볼트 바이오테라퓨틱스 인코퍼레이티드 Aminobenzazepine compounds, immunoconjugates, and uses thereof
AU2020359446A1 (en) 2019-09-30 2022-04-21 Bolt Biotherapeutics, Inc. Amide-linked, aminobenzazepine immunoconjugates, and uses thereof
MX2022004875A (en) 2019-10-25 2022-06-17 Bolt Biotherapeutics Inc Thienoazepine immunoconjugates, and uses thereof.
US20230139178A1 (en) 2020-03-12 2023-05-04 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
WO2021182574A1 (en) 2020-03-12 2021-09-16 東レ株式会社 Medicament for treatment and/or prevention of cancer
US20230129035A1 (en) 2020-03-12 2023-04-27 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
EP4119158A1 (en) 2020-03-12 2023-01-18 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
BR112022018171A2 (en) 2020-03-12 2022-10-25 Toray Industries MEDICINE FOR THE TREATMENT AND/OR PREVENTION OF CANCER, AGENTS ENHANCEMENT THE DRUG EFFECTIVENESS OF A PHARMACEUTICAL COMPOSITION AND METHOD TO TREAT AND/OR PREVENT CANCER
WO2021226440A1 (en) 2020-05-08 2021-11-11 Bolt Biotherapeutics, Inc. Elastase-substrate, peptide linker immunoconjugates, and uses thereof
EP4196168A1 (en) 2020-08-13 2023-06-21 Bolt Biotherapeutics, Inc. Pyrazoloazepine immunoconjugates, and uses thereof
CN117545508A (en) 2021-06-23 2024-02-09 东丽株式会社 Medicine for treating and/or preventing cancer
JPWO2022270524A1 (en) 2021-06-23 2022-12-29
CN117677398A (en) 2021-07-27 2024-03-08 东丽株式会社 Medicine for treating and/or preventing cancer
WO2023008461A1 (en) 2021-07-27 2023-02-02 東レ株式会社 Medicament for treatment and/or prevention of cancer
AU2022319362A1 (en) 2021-07-27 2024-02-29 Toray Industries, Inc. Medicament for treatment and/or prevention of cancer
CA3230737A1 (en) 2021-09-03 2023-03-09 Toray Industries, Inc. Pharmaceutical composition for cancer treatment and/or prevention
WO2023076599A1 (en) 2021-10-29 2023-05-04 Bolt Biotherapeutics, Inc. Tlr agonist immunoconjugates with cysteine-mutant antibodies, and uses thereof
WO2024005123A1 (en) * 2022-06-30 2024-01-04 東レ株式会社 Pharmaceutical composition for treating and/or preventing cancer

Family Cites Families (104)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DE69535243T2 (en) 1994-07-13 2007-05-10 Chugai Seiyaku K.K. AGAINST HUMAN INTERLEUKIN-8 DIRECTED, RECONSTITUTED HUMAN ANTIBODY
ATE449342T1 (en) 1994-09-19 2009-12-15 Ricardo J Moro DETECTION OF CANCER
US5698396A (en) 1995-06-07 1997-12-16 Ludwig Institute For Cancer Research Method for identifying auto-immunoreactive substances from a subject
FR2761994B1 (en) 1997-04-11 1999-06-18 Centre Nat Rech Scient PREPARATION OF MEMBRANE RECEPTORS FROM EXTRACELLULAR BACULOVIRUSES
TR200101541T2 (en) 1998-04-03 2002-06-21 Chugai Seiyaku Kabushiki Kaisha Humanized antibody against human tissue factor (TF).
DE69942021D1 (en) 1998-04-20 2010-04-01 Glycart Biotechnology Ag GLYCOSYLATION ENGINEERING OF ANTIBODIES TO IMPROVE ANTIBODY-DEPENDENT CELL-EMITTED CYTOTOXICITY
US20030118599A1 (en) 1999-04-02 2003-06-26 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
RU2234942C2 (en) 1998-07-14 2004-08-27 Корикса Корпорейшн Tumor polypeptide isolated from prostate and polynucleotide encoding thereof
HUP0203035A3 (en) 1998-07-14 2007-12-28 Corixa Corp Compositions and methods for therapy and diagnosis of prostate cancer
GB9815909D0 (en) 1998-07-21 1998-09-16 Btg Int Ltd Antibody preparation
CA2339045A1 (en) 1998-07-21 2000-02-03 Connex Gesellschaft Zur Optimierung Von Forschung Und Entwicklung Mbh Anti hepatitis c virus antibody and uses thereof
US6969518B2 (en) 1998-12-28 2005-11-29 Corixa Corporation Compositions and methods for the therapy and diagnosis of breast cancer
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
WO2000052204A2 (en) 1999-02-22 2000-09-08 Orntoft Torben F Gene expression in bladder tumors
WO2000060077A2 (en) 1999-04-02 2000-10-12 Corixa Corporation Compounds for therapy and diagnosis of lung cancer and methods for their use
US6444425B1 (en) 1999-04-02 2002-09-03 Corixa Corporation Compounds for therapy and diagnosis of lung cancer and methods for their use
DK2270147T4 (en) * 1999-04-09 2020-08-31 Kyowa Kirin Co Ltd METHOD OF MONITORING THE ACTIVITY OF IMMUNOLOGICAL FUNCTIONAL MOLECULE
US6949245B1 (en) 1999-06-25 2005-09-27 Genentech, Inc. Humanized anti-ErbB2 antibodies and treatment with anti-ErbB2 antibodies
JP2003528587A (en) 1999-10-29 2003-09-30 ヒューマン ジノーム サイエンシーズ, インコーポレイテッド 27 human secretory proteins
US20020006404A1 (en) 1999-11-08 2002-01-17 Idec Pharmaceuticals Corporation Treatment of cell malignancies using combination of B cell depleting antibody and immune modulating antibody related applications
WO2001072295A2 (en) 2000-03-29 2001-10-04 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
FR2807767B1 (en) 2000-04-12 2005-01-14 Lab Francais Du Fractionnement MONOCLONAL ANTIBODIES ANTI-D
US7919467B2 (en) 2000-12-04 2011-04-05 Immunotope, Inc. Cytotoxic T-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
US7408041B2 (en) 2000-12-08 2008-08-05 Alexion Pharmaceuticals, Inc. Polypeptides and antibodies derived from chronic lymphocytic leukemia cells and uses thereof
US20040029114A1 (en) 2001-01-24 2004-02-12 Eos Technology, Inc. Methods of diagnosis of breast cancer, compositions and methods of screening for modulators of breast cancer
RU2306952C2 (en) 2001-01-31 2007-09-27 Байоджен Айдек Инк. Method for treating the cases of malignant tumors containing b-cells with a combination of applications related to antibodies reducing b-cells number and with immunomodulating antibodies
AU2002311787A1 (en) 2001-03-28 2002-10-15 Zycos Inc. Translational profiling
WO2002083070A2 (en) 2001-04-10 2002-10-24 Corixa Corporation Compounds for immunotherapy and diagnosis of colon cancer and methods for their use
WO2002092001A2 (en) 2001-05-11 2002-11-21 Corixa Corporation Compositions and methods for the therapy and diagnosis of lung cancer
US20030190640A1 (en) 2001-05-31 2003-10-09 Mary Faris Genes expressed in prostate cancer
RU2319709C2 (en) 2001-07-17 2008-03-20 Рисерч Дивелопмент Фаундейшн Proapoptosis proteins-containing therapeutic agents
CA2454048C (en) 2001-07-17 2011-05-03 Research Development Foundation Therapeutic agents comprising pro-apoptotic proteins
US20040142325A1 (en) 2001-09-14 2004-07-22 Liat Mintz Methods and systems for annotating biomolecular sequences
CA2476518A1 (en) 2002-02-22 2003-09-04 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20050003390A1 (en) 2002-05-17 2005-01-06 Axenovich Sergey A. Targets for controlling cellular growth and for diagnostic methods
US20040126379A1 (en) 2002-08-21 2004-07-01 Boehringer Ingelheim International Gmbh Compositions and methods for treating cancer using cytotoxic CD44 antibody immunoconjugates and chemotherapeutic agents
EP2179742A1 (en) 2002-11-26 2010-04-28 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
WO2004076682A2 (en) 2003-02-26 2004-09-10 Surromed, Inc. Targets for controlling cellular growth and for diagnostic methods
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
WO2004097051A2 (en) 2003-04-29 2004-11-11 Wyeth Methods for diagnosing aml and mds differential gene expression
EP2481814A3 (en) 2003-06-09 2012-10-10 The Regents of the University of Michigan Compositions and methods for treating and diagnosing cancer
WO2005007830A2 (en) 2003-07-14 2005-01-27 Mayo Foundation For Medical Education And Research Methods and compositions for diagnosis, staging and prognosis of prostate cancer
US20050032113A1 (en) 2003-07-17 2005-02-10 Mitsubishi Pharma Corporation Membrane protein library for proteome analysis and method for preparing same
DE10359795A1 (en) 2003-12-19 2005-07-21 Bayer Technology Services Gmbh Tube evaporator device
JP2008506352A (en) 2004-01-26 2008-03-06 デビオビジョン・インコーポレーテッド Novel organism-specific polypeptides and uses thereof
DK1735348T3 (en) 2004-03-19 2012-07-16 Imclone Llc Human anti-epidermal growth factor receptor antibody
RU2429245C2 (en) 2004-03-30 2011-09-20 Глаксо Груп Лимитед Immunoglobulins
WO2005100998A2 (en) 2004-04-16 2005-10-27 Europroteome Ag Membrane markers for use in cancer diagnosis and therapy
WO2006002378A2 (en) 2004-06-23 2006-01-05 Avalon Pharmaceuticals Determining cancer-linked genes and therapeutic targets using molecular cytogenetic methods
RU2297241C2 (en) 2004-11-22 2007-04-20 Государственное учреждение Российский научный центр рентгенорадиологии Министерства здравоохранения и социального развития Chimeric protein for malignant lymphoma treatment
KR101285047B1 (en) 2005-01-19 2013-07-10 제리아 신야쿠 고교 가부시키 가이샤 Antitumor agent
CN101141981A (en) 2005-01-21 2008-03-12 健泰科生物技术公司 Fixed dosing of her antibodies
CA2598217A1 (en) 2005-02-18 2006-08-24 Marsha A. Moses Cyr61 as a biomarker for diagnosis and prognosis of cancers of epithelial origin
CA2602088C (en) 2005-03-11 2021-07-27 Ciphergen Biosystems, Inc. Biomarkers for ovarian cancer and endometrial cancer
JP2006318040A (en) 2005-05-10 2006-11-24 Ricoh Co Ltd Service providing system, program and recording medium
JP2006316040A (en) 2005-05-13 2006-11-24 Genentech Inc Herceptin(r) adjuvant treatment
US8014957B2 (en) 2005-12-15 2011-09-06 Fred Hutchinson Cancer Research Center Genes associated with progression and response in chronic myeloid leukemia and uses thereof
JP5490413B2 (en) 2006-01-05 2014-05-14 ジ・オハイオ・ステイト・ユニバーシティ・リサーチ・ファウンデイション Abnormal microRNA expression in pancreatic endocrine and acinar tumors
EP1991701A4 (en) 2006-02-14 2010-03-17 Dana Farber Cancer Inst Inc Compositions, kits, and methods for identification, assessment, prevention, and therapy of cancer
US20100015724A1 (en) 2006-08-17 2010-01-21 Peter Hornbeck Lysine acetylation sites
US20080107668A1 (en) 2006-08-30 2008-05-08 Immunotope, Inc. Cytotoxic t-lymphocyte-inducing immunogens for prevention, treatment, and diagnosis of cancer
US7615349B2 (en) 2006-09-07 2009-11-10 H. Lee Moffitt Cancer Center And Research Institute, Inc. Melanoma gene signature
WO2008059252A2 (en) 2006-11-15 2008-05-22 Oxford Biomedica (Uk) Limited Methods and composition fro t cell receptors which recognize 5t4 antigen
JP5439815B2 (en) 2007-10-25 2014-03-12 東レ株式会社 Cancer detection method
JP5663834B2 (en) 2008-03-14 2015-02-04 東ソー株式会社 Method for producing recombinant antibody
KR20210131473A (en) 2008-03-18 2021-11-02 제넨테크, 인크. Combinations of an anti-HER2 antibody-drug conjugate and chemotherapeutic agents, and methods of use
PL2837383T3 (en) 2008-08-05 2017-07-31 Toray Industries, Inc. Immunity-inducing agent
ES2570731T3 (en) 2008-08-05 2016-05-20 Toray Industries Cancer screening method
KR101667319B1 (en) 2008-08-05 2016-10-18 도레이 카부시키가이샤 Pharmaceutical composition for treatment and prevention of cancer
WO2010018883A1 (en) 2008-08-14 2010-02-18 Myungjoo Kwon Magnetic-piezoelectric combine sensor using piezoelectric single crystal
EP2445520A4 (en) 2009-06-22 2013-03-06 Medimmune Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
JP5734978B2 (en) * 2009-08-19 2015-06-17 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung Antibodies for integrin complex detection in FFPE materials
PL2480671T3 (en) 2009-09-22 2015-12-31 Probiogen Ag Process for producing molecules containing specialized glycan structures
CA2788720C (en) 2010-02-04 2019-08-20 Takayoshi Ido Medicament for treating and/or preventing cancer
US8709418B2 (en) 2010-02-04 2014-04-29 Toray Industries, Inc. Pharmaceutical composition for treating CAPRIN-1 expressing cancer
AU2011211682B2 (en) * 2010-02-04 2015-08-13 Toray Industries, Inc. Pharmaceutical composition for treating and/or preventing cancer
PL2532365T3 (en) 2010-02-04 2016-12-30 Pharmaceutical composition for treatment and/or prevention of cancer
PT2532366T (en) 2010-02-04 2016-12-20 Toray Industries Pharmaceutical composition for treating and/or preventing cancer
AU2011211699B2 (en) 2010-02-04 2015-01-22 Toray Industries, Inc. Pharmaceutical composition for treating and/or preventing cancer
EP2360965A1 (en) 2010-02-12 2011-08-24 ST-Ericsson SA Handling of measurements for selecting a cell in a network
MX341925B (en) 2010-03-29 2016-09-07 Zymeworks Inc Antibodies with enhanced or suppressed effector function.
WO2012005550A2 (en) 2010-07-08 2012-01-12 강원대학교산학협력단 Pharmaceutical composition for treating gallbladder carcinoma, and method for inhibiting growth and metastasis of gallbladder carcinoma and treating gallbladder carcinoma, using same
CN103261224B (en) 2010-07-26 2015-10-07 瑟维尔实验室 For the method and composition of liver cancer treatment
MX348581B (en) 2011-08-04 2017-06-20 Toray Industries Cancer treatment and/or prevention drug composition.
ES2609846T3 (en) 2011-08-04 2017-04-24 Toray Industries, Inc. Pharmaceutical composition for the treatment and / or prevention of pancreatic cancer
EP2740796B1 (en) 2011-08-04 2017-05-17 Toray Industries, Inc. Pharmaceutical composition for treatment and/or prophylaxis of cancer
DK2740795T3 (en) 2011-08-04 2017-01-16 Toray Industries Pharmaceutical composition for the treatment and / or prevention of cancer
WO2013018891A1 (en) 2011-08-04 2013-02-07 東レ株式会社 Pharmaceutical composition for treatment and/or prophylaxis of cancer
EP2740793B1 (en) 2011-08-04 2017-11-29 Toray Industries, Inc. Drug composition for cancer treatment and/or prevention
MX349907B (en) 2011-08-04 2017-08-18 Toray Industries Method for detecting pancreatic cancer.
JP5855969B2 (en) 2012-02-15 2016-02-09 株式会社神戸製鋼所 Metal plate bending method
DK2818481T3 (en) 2012-02-21 2019-10-14 Toray Industries Pharmaceutical composition for the treatment and / or prevention of cancer
KR102009238B1 (en) 2012-02-21 2019-08-09 도레이 카부시키가이샤 Pharmaceutical composition for treatment and/or prevention of cancer
RU2633505C2 (en) 2012-02-21 2017-10-12 Торэй Индастриз, Инк. Pharmaceutical composition for cancer treatment and/or prevention
PL2818483T3 (en) 2012-02-21 2018-01-31 Toray Industries Medicinal composition for treating and/or preventing cancer
EP2832368B1 (en) 2012-03-28 2021-11-10 Hiroshima University Fibrosis suppression by inhibiting integrin alpha8beta1 function
KR102155531B1 (en) 2012-03-30 2020-09-15 도레이 카부시키가이샤 Pharmaceutical composition for treatment and/or prevention of liver cancer
MX357965B (en) 2012-03-30 2018-08-01 Toray Industries Pharmaceutical composition for treatment and/or prevention of gall bladder cancer.
WO2013147189A1 (en) 2012-03-30 2013-10-03 積水化学工業株式会社 Curable resin composition for vapor deposition, resin protective film, oragnic optical device, and method for manufacturing organic optical device
CN104471404B (en) 2012-07-19 2017-03-01 东丽株式会社 The detection method of cancer
WO2014014082A1 (en) 2012-07-19 2014-01-23 東レ株式会社 Cancer detection method
WO2015020212A1 (en) 2013-08-09 2015-02-12 東レ株式会社 Pharmaceutical composition for treatment and/or prevention of cancer

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160208014A1 (en) * 2008-08-05 2016-07-21 Toray Industries, Inc. Pharmaceutical composition for treatment and prevention of cancers
US9982059B2 (en) * 2008-08-05 2018-05-29 Toray Industries, Inc. Pharmaceutical composition for treatment and prevention of cancers
US11137401B2 (en) 2008-08-05 2021-10-05 Toray Industries, Inc. Method for detecting cancer using CAPRIN-1 as a marker
US9796775B2 (en) 2011-08-04 2017-10-24 Toray Industries, Inc. Method for detecting pancreatic cancer
US9862774B2 (en) 2013-08-09 2018-01-09 Toray Industries, Inc. Pharmaceutical composition for treatment and/or prevention of cancer
US10675358B2 (en) 2016-07-07 2020-06-09 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11110178B2 (en) 2016-07-07 2021-09-07 The Board Of Trustees Of The Leland Standford Junior University Antibody adjuvant conjugates
US11547761B1 (en) 2016-07-07 2023-01-10 The Board Of Trustees Of The Leland Stanford Junior University Antibody adjuvant conjugates
US11400164B2 (en) 2019-03-15 2022-08-02 Bolt Biotherapeutics, Inc. Immunoconjugates targeting HER2

Also Published As

Publication number Publication date
AU2014303375A1 (en) 2016-03-03
AU2014303375B2 (en) 2019-07-04
RU2016107884A (en) 2017-09-14
CA2918989C (en) 2021-11-02
ES2704909T3 (en) 2019-03-20
KR102255616B1 (en) 2021-05-25
HUE042081T2 (en) 2019-06-28
EP3031826A1 (en) 2016-06-15
JP6447130B2 (en) 2019-01-09
EP3031826B1 (en) 2018-10-10
RU2678138C2 (en) 2019-01-23
BR112016001753B1 (en) 2023-10-03
JPWO2015020212A1 (en) 2017-03-02
BR112016001753A2 (en) 2017-08-29
MX360671B (en) 2018-11-13
PT3031826T (en) 2019-01-18
PL3031826T3 (en) 2019-03-29
TR201819812T4 (en) 2019-01-21
US9862774B2 (en) 2018-01-09
DK3031826T3 (en) 2018-12-17
MX2016001640A (en) 2016-06-02
CN105452294A (en) 2016-03-30
KR20160038892A (en) 2016-04-07
EP3031826A4 (en) 2017-03-29
CN105452294B (en) 2019-08-02
CA2918989A1 (en) 2015-02-12
WO2015020212A1 (en) 2015-02-12

Similar Documents

Publication Publication Date Title
US9862774B2 (en) Pharmaceutical composition for treatment and/or prevention of cancer
AU2012290955B2 (en) Drug composition for cancer treatment and/or prevention
AU2012290954B2 (en) Pharmaceutical composition for treatment and/or prophylaxis of cancer
US9181348B2 (en) Pharmaceutical composition for treatment and/or prophylaxis of cancer
US9260513B2 (en) Pharmaceutical composition for treatment and/or prevention of cancer
US9573993B2 (en) Pharmaceutical composition for treatment of cancer comprising an anti-CAPRIN-1 peptide antibody
US9273130B2 (en) Pharmaceutical composition for treatment and/or prevention of cancer
US9180188B2 (en) Pharmaceutical composition for treatment and/or prophylaxis of cancer
AU2013223147B2 (en) Pharmaceutical composition for treatment and/or prevention of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: TORAY INDUSTRIES, INC., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:OKANO, FUMIYOSHI;SAITO, TAKANORI;MINAMIDA, YOSHITAKA;AND OTHERS;SIGNING DATES FROM 20151201 TO 20151214;REEL/FRAME:037695/0124

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4